WO2018071919A1 - IL15/IL15Rα HETERODIMERIC FC-FUSION PROTEINS - Google Patents

IL15/IL15Rα HETERODIMERIC FC-FUSION PROTEINS Download PDF

Info

Publication number
WO2018071919A1
WO2018071919A1 PCT/US2017/056829 US2017056829W WO2018071919A1 WO 2018071919 A1 WO2018071919 A1 WO 2018071919A1 US 2017056829 W US2017056829 W US 2017056829W WO 2018071919 A1 WO2018071919 A1 WO 2018071919A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
protein
polypeptide sequence
domain
fusion protein
Prior art date
Application number
PCT/US2017/056829
Other languages
English (en)
French (fr)
Inventor
Matthew Bernett
Rumana RASHID
John Desjarlais
Rajat VARMA
Christine Bonzon
Original Assignee
Xencor, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP17804322.0A priority Critical patent/EP3526241A1/en
Priority to CN201780067629.XA priority patent/CN110214147A/zh
Priority to BR112019007281A priority patent/BR112019007281A2/pt
Priority to PE2019000823A priority patent/PE20191033A1/es
Priority to SG11201903304YA priority patent/SG11201903304YA/en
Priority to MX2019004328A priority patent/MX2019004328A/es
Priority to CA3040504A priority patent/CA3040504A1/en
Priority to KR1020247008957A priority patent/KR20240038176A/ko
Priority to RU2019114268A priority patent/RU2779938C2/ru
Priority to CR20230179A priority patent/CR20230179A/es
Priority to CR20190229A priority patent/CR20190229A/es
Priority to AU2017342560A priority patent/AU2017342560B2/en
Priority to JP2019519632A priority patent/JP7142630B2/ja
Priority to KR1020197012663A priority patent/KR102649972B1/ko
Application filed by Xencor, Inc. filed Critical Xencor, Inc.
Publication of WO2018071919A1 publication Critical patent/WO2018071919A1/en
Priority to PH12019500781A priority patent/PH12019500781A1/en
Priority to IL265998A priority patent/IL265998B1/en
Priority to ZA2019/02380A priority patent/ZA201902380B/en
Priority to CONC2019/0003943A priority patent/CO2019003943A2/es
Priority to AU2022203782A priority patent/AU2022203782B2/en
Priority to JP2022145394A priority patent/JP2022180450A/ja
Priority to AU2024203151A priority patent/AU2024203151A1/en
Priority to IL312989A priority patent/IL312989A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5443IL-15
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7155Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/35Valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/53Hinge
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"

Definitions

  • IL-2 and IL-15 function in aiding the proliferation and differentiation of B cells, T cells, and NK cells.
  • IL-2 is also essential for regulatory T cell (Treg) function and survival.
  • Both cytokines exert their cell signaling function through binding to a trimeric complex consisting of two shared receptors, the common gamma chain (yc; CD132) and IL-2 receptor B-chain (IL-2R
  • cytokines are considered as potentially valuable therapeutics in oncology and IL-2 has been approved for use in patients with metastatic renal-cell carcinoma and malignant melanoma.
  • IL-2 has been approved for use in patients with metastatic renal-cell carcinoma and malignant melanoma.
  • IL-15 Currently there are no approved uses of recombinant IL-15, although several clinical trials are ongoing.
  • IL-2 presents several challenges as a therapeutic agent. First, it preferentially activates T cells that express the high affinity receptor complex, which depends on CD25 expression. Because Treg cells constitutively express CD25, they compete for IL-2 supplies with effector T cells, whose activation is preferred for oncology treatment. This imbalance has led to the concept of high dose IL-2. However, this approach creates additional problems because of IL-2-mediated toxicities such as vascular leak syndrome.
  • IL-2 is secreted primarily by activated T cells, while its receptors are located on activated T cells, Tregs, NK cells, and B cells.
  • IL-15 is produced on monocytes and dendritic cells and is primarily presented as a membrane-bound heterodimeric complex with IL-15R present on the same cells. Its effects are realized through trans-presentation of the IL-15/IL-15R complex to NK cells and CD8+ T cells expressing IL-2R
  • the present invention provides a heterodimeric protein comprising a) a first fusion protein comprising a first protein domain and a first Fc domain, wherein the first protein domain is covalently attached to the N-terminus of the first Fc domain using a first domain linker; b) a second fusion protein comprising a second protein domain and a second Fc domain, wherein the second protein domain is covalently attached to the N-terminus of the Fc domain using a seconddomain linker; wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K L368D/K370S : S267K/LS364K E357Q; S364K E357Q : L368D/K370S; L368D/ 370S : S364K; L368E/ 370S : S364K; T411T/E360E/Q362E : D401K;
  • the first protein domain is covalently attached to the N-terminus of the first Fc domain directly and without using the first domain linker and/or the second protein domain is covalently attached to the N-terminus of the second Fc domain directly and without using the second domain linker.
  • the heterodimeric protein comprises: (i) the first fusion protein having a polypeptide sequence of SEQ ID NO:XX (XENP15902) and the second fusion protein having a polypeptide sequence of SEQ ID NO:XX (XENP5908), (ii) the first fusion protein having a polypeptide sequence of SEQ ID NO:XX (XENP15902) and the second fusion protein having a polypeptide sequence of SEQ ID NO:XX (XENP15909), (iii) the first fusion protein having a polypeptide sequence of SEQ ID NO:XX (XENP16479) and the second fusion protein having a polypeptide sequence of SEQ ID NO:XX (XENP15908), (iv) the first fusion protein having a polypeptide sequence of SEQ ID NO:XX (XENP15902) and the second fusion protein having a polypeptide sequence of SEQ ID NO:XXX
  • the heterodimeric protein is selected from the group consisting of XENP20818, XENP20819, XENP21471, XENP21472, XENP21473, XENP21474, XENP21475, XENP21476, XENP21477, XENP22013, XENP22815, XENP22816, XENP22817, XENP22818, XENP22819, XENP22820, XENP22821, XENP22822, XENP22823, XENP22824, XENP22825, XENP22826, XENP22827, XENP22828, XENP22829, XENP22830, XENP22831, XENP22832, XENP22833, XENP22834, XENP23343, XEN
  • the invention provides a heterodimeric protein comprising: a) a fusion protein comprising a first protein domain, a second protein domain, and a first Fc domain, wherein the first protein domain is covalently attached to the N-terminus of the second protein domain using a first domain linker, and wherein the second protein domain is covalently attached to the N-terminus of the first Fc domain using a second domain linker; b) a second Fc domain; wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K/L368D/K370S :
  • the first fusion protein has a polypeptide sequence of SEQ ID NO:XX (16478) and the Fc domain has a polypeptide sequence of SEQ ID NO:XX (8924).
  • the heterodimeric protein can be XENP21478.
  • the invention provides a heterodimeric protein comprising: a) a fusion protein comprising a first protein domain and a first Fc domain, wherein the first protein domain is covalently attached to the N-terminus of the first Fc domain using a domain linker; b) a second Fc domain; and c) a second protein domain noncovalently attached to the first protein domain; wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K/L368D/K370S : S267K/LS364K/E357Q; S364K/E357Q : L368D/ 370S; L368D/K370S : S364K; L368E/ 370S : S364K; T411T E360E/Q362E : D401K; L368D/K370S : S364K E357L and K370S : S36
  • the heterodimer protein comprises: (i) the fusion protein having a polypeptide sequence of SEQ ID NO:XX (XENP16481), the second Fc domain having a polypeptide sequence of SEQ ID NO:XX (8793), and a second protein domain having a polypeptide sequence of SEQ ID NO:XX (16484); (ii) the fusion protein having a polypeptide sequence of SEQ ID NO:XX (17034), the second Fc domain having a
  • polypeptide sequence of SEQ ID NO:XX (8793), and a second protein domain having a polypeptide sequence of SEQ ID NO:XX (16484); (iii) the fusion protein having a
  • polypeptide sequence of SEQ ID NO:XX (8793), and a second protein domain having a polypeptide sequence of SEQ ID NO:XX (16484); (iv) the fusion protein having a
  • polypeptide sequence of SEQ ID NO:XX (8793), and a second protein domain having a polypeptide sequence of SEQ ID NO:XX (17083); or (xiv) the fusion protein having a polypeptide sequence of SEQ ID NO:XX (15908), the second Fc domain having a
  • the heterodimer protein can be selected from the group consisting of XENP21479, XENP22357, XENP22354, XENP22355,
  • the invention provides a heterodimeric protein comprising: a) a first fusion protein comprising a first protein domain and a first Fc domain, wherein the first protein domain is covalently attached to the N-terminus of said first Fc domain using a domain linker; b) a second fusion protein comprising a second heavy chain comprising a second protein domain and a first second heavy chain comprising a second Fc domain, wherein the second protein domain is covalently attached to the C-terminus of the second Fc domain using a domain linker; c) a third protein domain noncovalently attached to the first protein domain of the first fusion protein; and d) a fourth protein domain noncovalently attached to the second protein domain of the second fusion protein, wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K L368D/K370S : S267K/LS364K/E357Q; S364
  • the heterodimer protein comprises (i) the first fusion protein has a polypeptide sequence of SEQ ID NO:XX (17023) the second fusion protein has a polypeptide sequence of SEQ ID NO:XX (17023), the third protein domain has a polypeptide sequence of SEQ ID NO:XX (16484), and the fourth protein domain has a polypeptide sequence of SEQ ID NO:XX (16484) or (ii) the first fusion protein has a polypeptide sequence of SEQ ID NO:XX (17581), the second fusion protein has a
  • the heterodimer protein can be XENP21978 or XENP22634.
  • the invention provides a heterodimeric protein comprising: a) a first fusion protein comprising a first Fc domain and a first protein domain, wherein the first Fc domain is covalently attached to the N-terminus of the first protein domain using a domain linker; b) a second Fc domain, and c) a second protein domain noncovalently attached to the first protein domain of the first fusion protein; wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K L368D/K370S : S267K/LS364K/E357Q; S364K/E357Q :
  • the heterodimer protein comprises (i) the first fusion protein having a polypeptide sequence of SEQ ID NO: XX (17603), the second Fc domain having a polypeptide sequence of SEQ ID NO: XX (8927), and the second protein domain having a polypeptide sequence of SEQ ID NO: XX (16484); or ii) the first fusion protein having a polypeptide sequence of SEQ ID NO: XX (17605), the second Fc domain having a polypeptide sequence of SEQ ID NO: XX (8927), and the second protein domain having a polypeptide sequence of SEQ ID NO: XX (17074).
  • the first and/or the second Fc domains can have an additional set of amino acid substitutions comprising
  • the first and/or the second Fc domains have an additional set of amino acid substitutions consisting of G236R/L328R, E233P L234V/L235A/G236del/S239K, E233P/L234V L235A/G236del/S267K, E233P/L234V/L235A/G236del/S239K/A327G, E233P/L234V/L235A/G236del/S267K/A327G and E233P/L234V L235A/G236del, according to EU numbering.
  • the IL15 protein has a polypeptide sequence selected from the group consisting of SEQ ID NO:l (full-length human IL15) and SEQ ID NO:2 (truncated human IL15), and the IL15R protein has a polypeptide sequence selected from the group consisting of SEQ ID NO:3 (full-length human IL15R ) and SEQ ID NO:4 (sushi domain of human IL15R ).
  • the IL15 protein and the IL15R protein have a set of amino acid substitutions or additions selected from the group consisting of E87C : D96/P97/C98; E87C : D96/C97/A98; V49C : S40C; L52C : S40C; E89C : K34C; Q48C : G38C; E53C : L42C; C42S : A37C; and L45C : A37Q respectively.
  • the present invention provides a heterodimeric protein selected from the group consisting of XENP20818, XENP20819, XENP21471, XENP21472, XENP21473, XENP21474, XENP21475, XENP21476, XENP21477, XENP21478, XENP21479, XENP21978, XENP22013, XENP22015, XENP22017, XENP22354, XENP22355, XENP22356, XENP22357, XENP22358, XENP22359, XENP22360, XENP22361, XENP22362, XENP22363, XENP22364, XENP22365, XENP22366, XENP22637, and XENP22639.
  • the present invention provides a heterodimeric protein selected from the group consisting of XENP20818, XENP20819, XENP21471, XENP21472, XENP21473, XENP21474, XENP21475, XENP21476, XENP21477, XENP22013, XENP22815, XENP22816, XENP22817, XENP22818, XENP22819, XENP22820, XENP22821, XENP22822, XENP22823, XENP22824, XENP22825, XENP22826, XENP22827, XENP22828, XENP22829, XENP22830, XENP22831, XENP22832, XENP22833, XENP22834, XENP23343, XEN
  • Figure 1 depicts the structure of IL-15 in complex with its receptors IL-15R (CD215), IL-15RP (CD122), and the common gamma chain (CD132).
  • Figures 2A-2B depict the sequences for IL-15 and its receptors.
  • Figure 2A shows the sequences for human IL-15, human IL-15R and human IL-15R
  • Figure 2A shows the sequences for the human common gamma receptor.
  • Figures 3A-3E depict useful pairs of Fc heterodimerization variant sets
  • FIG. 3D and 3E there are variants for which there are no corresponding "monomer 2" variants; these are pi variants which can be used alone on either monomer.
  • Figure 4 depict a list of isosteric variant antibody constant regions and their respective substitutions. pl_(-) indicates lower pi variants, while pl_(+) indicates higher pi variants. These can be optionally and independently combined with other
  • heterodimerization variants of the inventions (and other variant types as well, as outlined herein).
  • Figure 5 depict useful ablation variants that ablate FcyR binding (sometimes referred to as “knock outs” or “KO” variants). Generally, ablation variants are found on both monomers, although in some cases they may be on only one monomer.
  • Figures 6A-6E show a particularly useful embodiments of "non-cytokine" components of the invention.
  • Figure 7 depicts a number of exemplary variable length linkers.
  • these linkers find use linking the C-terminus of IL-15 and/or IL-15R (sushi) to the N-terminus of the Fc region.
  • these linkers find use fusing IL-15 to the IL-15R (sushi).
  • Figures 8A-8E show the sequences of several useful IL-15/R -Fc format backbones based on human IgGl, without the cytokine sequences (e.g., the 11-15 and/or IL- 15R (sushi)).
  • Backbone 1 is based on human IgGl (356E/358M allotype), and includes C220S on both chain, the S364K E357Q : L368D/K370S skew variants, the
  • Backbone 2 is based on human IgGl (356E/358M allotype), and includes C220S on both chain, the S364K : L368D/ 370S skew variants, the Q295E/N384D/Q418E/N421D pi variants on the chain with L368D/ 370S skew variants and the E233P/L234V/L235A/G236del/S267K ablation variants on both chains.
  • Backbone 3 is based on human IgGl (356E/358M allotype), and includes C220S on both chain, the S364K : L368E/K370S skew variants, the
  • Backbone 4 is based on human IgGl (356E/358M allotype), and includes C220S on both chain, the D401K : K360E/Q362E/T411E skew variants, the Q295E/N384D/Q418E/N421D pi variants on the chain with K360E/Q362E T411E skew variants and the E233P/L234V/L235A/G236del/S267K ablation variants on both chains.
  • Backbone 5 is based on human IgGl (356D/358L allotype), and includes C220S on both chain, the S364K/E357Q : L368D/K370S skew variants, the Q295E/N384D/Q418E/N421D pi variants on the chain with L368D/ 370S skew variants and the E233P L234V L235A/G236del/S267K ablation variants on both chains.
  • Backbone 6 is based on human IgGl (356E/358M allotype), and includes C220S on both chain, the
  • S364K E357Q L368D/K370S skew variants, Q295E/N384D/Q418E/N421D pi variants on the chain with L368D/K370S skew variants and the E233P L234V/L235A/G236del/S267K ablation variants on both chains, as well as an N297A variant on both chains.
  • Backbone 7 is identical to 6 except the mutation is N297S.
  • Alternative formats for backbones 6 and 7 can exclude the ablation variants E233P L234V L235A/G236del/S267K in both chains.
  • Backbone 8 is based on human IgG4, and includes the S364K/E357Q : L368D/K370S skew variants, the Q295E/N384D/Q418E/N421D pi variants on the chain with L368D/ 370S skew variants, as well as a S228P (EU numbering, this is S241P in Kabat) variant on both chains that ablates Fab arm exchange as is known in the art.
  • S228P EU numbering, this is S241P in Kabat
  • Backbone 9 is based on human IgG2, and includes the S364K/E357Q : L368D/K370S skew variants, the Q295E/N384D/Q418E/ 421D pi variants on the chain with L368D/K370S skew variants.
  • Backbone 10 is based on human IgG2, and includes the S364K E357Q : L368D/K370S skew variants, the Q295E/ 384D/Q418E/ 421D pi variants on the chain with L368D/K370S skew variants as well as a S267K variant on both chains.
  • Backbone 11 is identical to backbone 1, except it includes M428L/N434S Xtend mutations.
  • Backbone 12 is based on human IgGl (356E/358M allotype), and includes C220S on both identical chain, the the E233P/L234V/L235A/G236del/S267K ablation variants on both identical chains.
  • Backbone 13 is based on human IgGl (356E/358M allotype), and includes C220S on both chain, the S364K/E357Q : L368D/K370S skew variants, the
  • any IL-15 and IL-15R (sushi) pairs outlined herein including but not limited to IL-15/R -heteroFc, ncIL-15/R , scIL-15/R , and dsIL-15/R as schematically depicted in Figures 9A-9G, and Figures 39A-39D. Additionally, any IL-15 and/or IL- 15R (sushi) variants can be incorporated into these Figures 8A-8E backbones in any combination.
  • each of these backbones includes sequences that are 90%, 95%, 98% and 99% identical (as defined herein) to the recited sequences, and/or contain from 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 additional amino acid substitutions (as compared to the "parent" of the Figure, which, as will be appreciated by those in the art, already contain a number of amino acid modifications as compared to the parental human IgGl (or IgG2 or IgG4, depending on the backbone). That is, the recited backbones may contain additional amino acid
  • Figures 9A-9G depict several formats for the IL-15/R -Fc fusion proteins of the present invention.
  • IL-15R Heterodimeric Fc fusion or "IL-15/R -heteroFc" comprises IL-15 recombinantly fused to one side of a heterodimeric Fc and IL-15R (sushi) recombinantly fused to the other side of a heterodimeric Fc.
  • the IL-15 and IL-15R (sushi) may have a variable length Gly-Ser linker between the C-terminus and the N-terminus of the Fc region.
  • Single-chain IL-15/R -Fc fusion or "scIL-15/R -Fc" comprises IL- 15R (sushi) fused to IL-15 by a variable length linker (termed a "single-chain” IL-15/IL- 15R (sushi) complex or "scIL-15/R ”) which is then fused to the N-terminus of a heterodimeric Fc-region, with the other side of the molecule being "Fc-only" or "empty Fc".
  • Non-covalent IL-15/R -Fc or "ncIL-15/R -Fc" comprises IL-15R (sushi) fused to a heterodimeric Fc region, while IL-15 is transfected separatedly so that a non-covalent IL- 15/R complex is formed, with the other side of the molecule being "Fc-only” or "empty Fc".
  • Bivalent non-covalent IL-15/R -Fc fusion or "bivalent ncIL-15/R -Fc" comprises IL-15R (sushi) fused to the N-terminus of a homodimeric Fc region, while IL-15 is transfected separately so that a non-covalent IL-15/R complex is formed.
  • Bivalent single- chain IL-15/R -Fc fusion or "bivalent scIL-15/R -Fc" comprises IL-15 fused to IL-15R (sushi) by a variable length linker (termed a "single-chain” IL-15/IL-15R (sushi) complex or "scIL-15/R ”) which is then fused to the N-terminus of a homodimeric Fc- region.
  • Fc-non-covalent IL-15/R fusion or "Fc-ncIL-15/R” comprises IL- 15R (sushi) fused to the C-terminus of a heterodimeric Fc region, while IL-15 is transfected separately so that a non-covalent IL-15/R complex is formed, with the other side of the molecule being "Fc-only” or "empty Fc".
  • Fc-single-chain IL-15/R fusion or "Fc-scIL-15/R” comprises IL-15 fused to IL-15R (sushi) by a variable length linker (termed a "single-chain” IL-15/IL-15R (sushi) complex or "scIL-15/R ") which is then fused to the C- terminus of a heterodimeric Fc region, with the other side of the molecule being "Fc-only” or "empty Fc".
  • Figure 10 depicts sequences of XENP20818 and XENP21475, illustrative IL- 15/R -Fc fusion proteins of the "IL-15/R -heteroFc" format, with additional sequences being listed as XENPs 20819, 21471, 21472, 21473, 21474, 21476, and 21477 in the sequence listing.
  • IL-15 and IL-15R are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7), and slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 11 depicts sequences of XENP21478, an illustrative IL-15/R -Fc fusion protein of the "scIL-15/R -Fc" format, with additional sequences being listed as XENPs 21993, 21994, 21995, 23174, 23175, 24477, and 24480 in the sequence listing.
  • IL-15 and IL- 15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figures 12A-12B depict sequences of XENP21479, XENP22366 and XENP24348, illustrative IL-15/R -Fc fusion proteins of the "ncIL-15/R -Fc" format.
  • IL-15 and IL- 15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 13 depicts sequences of XENP21978, an illustrative IL-15/R -Fc fusion protein of the "bivalent ncIL-15/R -Fc" format, with additional sequences being listed as XENP21979 in the sequence listing.
  • IL-15 and IL-15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 14 depicts sequences of an illustrative IL-15/R -Fc fusion protein of the "bivalent scIL-15/R -Fc" format.
  • IL-15 and IL-15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 15 depicts sequences of XENP22637, an illustrative IL-15/R -Fc fusion protein of the "Fc-ncIL-15/R " format, with additional sequences being listed as XENP22638 in the sequence listing.
  • IL-15 and IL-15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 16 depicts sequences of an illustrative IL-15/R -Fc fusion protein of the "Fc-scIL-15/R " format.
  • IL-15 and IL-15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figures 17A-17E depict A) the IL-15/R -Fc fusion protein format for XENP20818, the purity and homogeneity of XENP20818 as determined by B) SEC and C) CEF, D) the affinity of XENP20818 for IL-2Rp as determined by Octet, and E) the stability of XENP20818 as determined by DSF
  • Figures 18A-18E depict A) the IL-15/R -Fc fusion protein format for XENP21478, the purity and homogeneity of XENP21478 as determined by B) SEC and C) CEF, D) the affinity of XENP21478 for IL-2Rp as determined by Octet, and E) the stability of XENP21478 as determined by DSF.
  • Figures 19A-19E depicts A) the IL-15/R -Fc fusion protein format for
  • XENP21479 the purity and homogeneity of XENP21479 as determined by B) SEC and C) CEF, D) the affinity of XENP21479 for IL-2Rp as determined by Octet, and E) the stability of XENP21479 as determined by DSF.
  • Figures 20A-20C depict the induction of A) NK (CD56+/CD16+) cells, B) CD4+ T cells, and C) CD8+ T cells proliferation by illustrative IL-15/R -Fc fusion proteins of the IL- 15/R -heteroFc format with different linker lengths based on Ki67 expression as measured by FACS.
  • Figures 21A-21C depict the induction of A) NK (CD56+/CD16+) cells, B) CD4+ T cells, and C) CD8+ T cells proliferation by illustrative IL-15/R -Fc fusion proteins of the scIL- 15/R -Fc format (XENP21478) and the ncIL-15/R -Fc format (XENP21479) based on Ki67 expression as measured by FACS.
  • Figure 22 depicts enhancement of IL-2 secretion by illustrative IL-15/R -Fc fusion proteins, an isotype control, and a bivalent anti-PD-1 antibody over PBS control in an SEB-stimulated PBMC assay.
  • Figure 23 depicts the survival curve for PBMC-engraf ted NSG mice following treatment with XENP20818 and recombinant IL-15.
  • Figure 24 depicts the concentration of IFNy in serum of NSG mice on Day 7 after engraf tment with human PBMCs and treatment with XENP20818 at the indicated concentrations.
  • Figures 25A-25C depict A) CD4+ T cell, B) CD8+ T cell, and C) CD45+ cell counts in whole blood of human PBMC-engraf ted NSG mice 7 days after treatment with
  • Figure 26 depicts a structural model of the IL-15/R heterodimer showing locations of engineered disulfide bond pairs.
  • Figure 27 depicts sequences for illustrative IL-15R (sushi) variants engineered with additional residues at the C-terminus to serve as a scaffold for engineering cysteine residues.
  • Figure 28 depicts sequences for illustrative IL-15 variants engineered with cysteines in order to form covalent disulfide bonds with IL-15R (sushi) variants engineered with cysteines.
  • Figure 29 depicts sequences for illustrative IL-15R (sushi) variants engineered with cysteines in order to form covalent disulfide bonds with IL-15 variants engineered with cysteines.
  • Figures 30A-30C depict IL-15/R heterodimers with and without engineered disulfide bonds between IL-15 and IL-15R (sushi).
  • Non-covalent IL-15/R heterodimer or "ncIL-15/R heterodimer” comprises IL-15R (sushi) and IL-15 transfected separately and non-covalently linked.
  • Disulfide-bonded IL-15/R heterodimer or "dsIL- 15/R heterodimer” ( Figure 30B) comprises IL-15R (sushi) and IL-15 transfected separately and covalently linked as a result of engineered cysteines.
  • Single-chain IL-15/R heterodimer or "scIL-15/R Heterodimer” comprises IL-15R (sushi) fused to IL-15 by a variable length Gly-Ser linker.
  • Figure 31 depicts sequences of XENP21996, an illustrative ncIL-15/R
  • Figure 32 depicts sequences of XENP22004, XENP22005, XENP22006,
  • XENP22008, and XENP22494 illustrative dsIL-15/R heterodimers, with additional sequences depicted as XENPs 22007, 22009, 22010, 22011, 22012, and 22493 in the sequence listing. It is important to note that these sequences were generated using polyhistidine (Hisx6 or HHHHHH) C-terminal tags at the C-terminus of IL-15R (sushi).
  • Figure 33 depicts the sequence for XENP22049, an illustrative scIL-15/R
  • Figure 35 depicts the purity and homogeneity of illustrative IL-15/R
  • Figure 36 depicts the stability and melting temperatures of illustrative IL-15/R heterodimers with and without engineered disulfide bonds as indicated by melting curves from DSF.
  • Figure 37 depicts the stability and melting temperatures of illustrative IL-15/R heterodimers with and without engineered disulfide bonds as indicated by melting curves from DSF.
  • Figure 38 depicts the expression yield, molecular weight, predicted change in affinity between IL-15 and IL-15R (sushi) as calculated by MOE software, melting temperature, and affinity for IL-2Rfi for IL-15/R heterodimers with and without engineered disulfide bonds. Mutations are indicated in parentheses after the relevant monomer.
  • Figures 39A-39D depict additional formats for the IL-15/R -Fc fusion proteins of the present invention with engineered disulfide bonds.
  • Disulfide-bonded IL-15/R heterodimeric Fc fusion or "dsIL-15/R -heteroFc" ( Figure 39A) is the same as "IL-15/Rct- heteroFc", but wherein IL-15R (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Disulfide-bonded IL-15/R Fc fusion or "dsIL-15/R -Fc" ( Figure 39B) is the same as "ncIL-15/R -Fc", but wherein IL-15R (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • bivalent dsIL-15/R -Fc ( Figure 39C) is the same as “bivalent ncIL-15/R -Fc", but wherein IL-15R (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Fc- disulfide-bonded IL-15/R fusion or “Fc-dsIL-15/R " ( Figure 39D) is the same as "Fc-ncIL- 15/R ", but wherein IL-15R (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Figures 40A-40B depicts sequences of XENP22013, XENP22014, XENP22015, and XENP22017, illustrative IL-15/R -Fc fusion protein of the "dsIL-15/R -heteroFc" format.
  • IL- 15 and IL-15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figures 41A-41B depict sequences of XENP22357, XENP22358, XENP22359, XENP22684, and XENP22361, iUustrative IL-15/R -Fc fusion proteins of the "dsIL-15/Ra-Fc" format. Additional sequences are depicted as XENPs 22360, 22362, 22363, 22364, 22365, and 22366 in the sequence listing.
  • IL-15 and IL-15R are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7), and slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 42 depicts sequences of XENP22634, XENP22635, and XENP22636, illustrative IL-15/R -Fc fusion proteins of the "bivalent dsIL-15/R -Fc" format. Additional sequences are depicted as XENP22687 in the sequence listing. IL-15 and IL-15R (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7), and slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 43 depicts sequences of XENP22639 and XENP22640, illustrative IL- 15/R -Fc fusion proteins of the "Fc-dsIL-15/R " format.
  • IL-15 and IL-15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 44 depicts the purity and homogeneity of illustrative IL-15/R -Fc fusion proteins with and without engineered disulfide bonds as determined by CEF.
  • Figures 45A-45C depict the induction of A) NK (CD56+/CD16+) cell, B) CD8+ T cell, and C) CD4+ T cell proliferation by illustrative IL-15/R -Fc fusion proteins with and without engineered disulfide bonds based on Ki67 expression as measured by FACS.
  • Figure 46 depicts the structure of IL-15 complexed with IL-15R , IL-2Rfi, and common gamma chain. Locations of substitutions designed to reduce potency are shown.
  • Figures 47A-47C depict sequences for illustrative IL-15 variants engineered for reduced potency. Included within each of these variant IL-15 sequences are sequences that are 90%, 95%, 98%, and 99% identical (as defined herein) to the recited sequences, and/or contain from 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 additional amino acid substitutions. In a non-limiting example, the recited sequences may contain additional amino acid modifications such as those contributing to formation of covalent disulfide bonds as described in Example 2.
  • Figures 48A-48D depict sequences of XENP22821, XENP22822, XENP23554, XENP23557, XENP23561, XENP24018, XENP24019, XENP24045, XENP24051, and
  • XENP24052 illustrative IL-15/R -Fc fusion proteins of the "IL-15/Ra-heteroFc" format engineered for lower potency. Additional sequences are depicted as XENPs 22815, 22816, 22817, 22818, 22819, 22820, 22823, 22824, 22825, 22826, 22827, 22828, 22829, 22830, 22831, 22832, 22833, 22834, 23555, 23559, 23560, 24017, 24020, 24043, and 24048 in the sequence listing.
  • IL-15 and IL-15R are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7), and slashes (/) indicate the border(s) between IL-15, IL- 15R , linkers, and Fc regions..
  • Figures 49A-49C depict sequences of XENP24015, XENP24050, XENP24475, XENP24476, XENP24478, XENP24479, and XENP24481, illustrative IL-15/R -Fc fusion proteins of the "scIL-15/R -Fc" format engineered for lower potency. Additional sequences are depicted as XENPs 24013, 24014, and 24016 in the sequence listing.
  • IL-15 and IL- 15R are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7), and slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figures 50 A-50B depict sequences of XENP24349, XENP24890, and XENP25138, illustrative IL-15/R -Fc fusion proteins of the "ncIL-15/R -Fc" format engineered for lower potency.
  • IL-15 and IL-15R are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7), and slashes (/) indicate the border(s) between IL-15, IL- 15R , linkers, and Fc regions.
  • Figure 51 depicts sequences of XENP22801 and XENP22802, illustrative ncIL- 15/R heterodimers engineered for lower potency. Additional sequences are depicted as XENPs 22791, 22792, 22793, 22794, 22795, 22796, 22803, 22804, 22805, 22806, 22807, 22808, 22809, 22810, 22811, 22812, 22813, and 22814 in the sequence listing. It is important to note that these sequences were generated using polyhistidine (Hisx6 or HHHHHH) C-terminal tags at the C-terminus of IL-15R (sushi)..
  • polyhistidine Hisx6 or HHHHHH
  • Figure 52 depicts sequences of XENP24342, an illustrative IL-15/R -Fc fusion protein of the "bivalent ncIL-15/R -Fc" format engineered for lower potency.
  • IL-15 and IL- 15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 53 depicts sequences of XENP23472 and XENP23473, illustrative IL- 15/R -Fc fusion proteins of the "dsIL-15/R -Fc" format engineered for lower potency.
  • IL-15 and IL-15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figures 54A-54C depict the induction of (A) NK cell, (B) CD8+ (CD45RA-) T cell, and (C) CD4+ (CD45RA-) T cell proliferation by variant IL-15/R -Fc fusion proteins based on Ki67 expression as measured by FACS.
  • Figure 55 depicts EC50 for induction of NK and CD8+ T cells proliferation by variant IL-15/R -Fc fusion proteins, and fold reduction in EC50 relative to XENP20818.
  • Figures 56A-56C depict the gating of lymphocytes and subpopulations for the experiments depicted in Figures 59A-59D.
  • Figure 56A shows the gated lymphocyte population.
  • Figure 56B shows the CD3-negative and CD3-positive subpopulations.
  • Figures 57A-57C depict the gating of CD3+ lymphocyte subpopulations for the experiments depicted in Figures 59A-59D.
  • Figure 57A shows the CD4+, CD8+ and ⁇ T cell subpopulations of the CD3+ T cells.
  • Figure 57B shows the CD45RA(-) and CD45RA(+) subpopulations of the CD4+ T cells.
  • Figure 57C shows the CD45RA(-) and CD45RA(+) subpopulation s of the CD8+ T cells.
  • Figures 58A-58B depict CD69 and CD25 expression before ( Figure 58A) and after ( Figure 58B) incubation of human PBMCs with XENP22821.
  • Figures 59A-59D depict cell proliferation in human PBMCs incubated for four days with the indicated variant IL-15/R -Fc fusion proteins.
  • Figures 59A-C show the percentage of proliferating NK cells (CD3-CD16+) ( Figure 59 A), CD8+ T cells
  • Figure 59D shows the fold change in EC50 of various IL15/IL15R Fc heterodimers relative to control (XENP20818).
  • Figures 60A-60D depict cell proliferation in human PBMCs incubated for three days with the indicated variant IL-15/R -Fc fusion proteins.
  • Figures 60A-C show the percentage of proliferating CD8+ (CD45RA-) T cells ( Figure A), CD4+ (CD45RA-) T cells ( Figure 60B), ⁇ T cells ( Figure 60C), and NK cells ( Figure 60D).
  • Figures 61A-61C depict the percentage of Ki67 expression on (A) CD8+ T cells, (B) CD4+ T cells, and (C) NK cells following treatment with additional IL-15/R variants.
  • Figures 62A-62E depict the percentage of Ki67 expression on (A) CD8+
  • CD45RA- T cells
  • B CD4+ (CD45RA-) T cells
  • C ⁇ T cells
  • D NK (CD16+CD8 -) cells
  • E NK (CD56+CD8 -) cells following treatment with IL-15/R variants.
  • Figures 63A-63E depict the percentage of Ki67 expression on (A) CD8+
  • CD45RA- T cells
  • B CD4+ (CD45RA-) T cells
  • C ⁇ T cells
  • D NK (CD16+CD8 -) cells
  • E NK (CD56+CD8 -) cells following treatment with IL-15/R variants.
  • Figures 64A-64D depict the percentage of Ki67 expression on (A) CD8+ T cells, (B) CD4+ T cells, (C) ⁇ T cells and (D) NK (CD16+) cells following treatment with additional IL-15/R variants engineered for decreased potency with different linker lengths.
  • Figures 65A-65D depict the percentage of Ki67 expression on (A) CD8+ T cells,
  • Figures 66A-66D depict gating of lymphocytes and subpopulations thereof for the experiments depicted in Figure 67.
  • Figure 66A shows gating of the lymphocyte population.
  • Figure 66B shows CD4+ and CD8+ T cells.
  • Figure 66C shows the CD45RA and CD27 expressing subpopulations of CD4+ T cells.
  • Figure 66D shows the CD45RA and CD27 expressing subpopulations of CD8+ T cells.
  • Figures 67A-67C depict STAT5 phosphorylation on A) CD8+ T cells (CD45RA- CD27-) and B) CD4+ T cells (CD45RA-CD27-) following incubation of PBMCs for 4 days with the indicated variant IL15/IL15R -Fc fusion proteins at the indicated concentrations.
  • Figure 67C shows the fold change in EC50 of various IL15/IL15R Fc heterodimers relative to control (XENP20818).
  • Figure 68 depicts IV-TV Dose PK of various IL-15/R -Fc fusion proteins or controls in C57BL/6 mice at 0.1 mg/kg single dose.
  • Figure 69 depicts the correlation of half-life vs NK cell potency.
  • Figure 70 shows that CD45+ cell levels are predictive of disease.
  • Figures 71A-71B depict the enhancement of engraftment by variant IL-15/R -Fc fusion proteins as indicated by CD45+ cell counts on Days A) 4 and B) 8.
  • Figures 72A-72C depict IFNy levels on Days (A) 4, (B) 7 and (C) 11 after treatment of NSG mice engrafted with human PBMCs with the indicated variant IL15/R -Fc fusion proteins or control.
  • Figures 73A-73C depict CD45+ lymphocyte cell counts on Days (A) 4, (B) 7, and
  • Figures 74A-74C depict NK cell (CD16+CD56+CD45RA+) counts on Days A) 4, B) 7 and C) 11 after treatment of NSG mice engrafted with human PBMCs with the indicated IL15/R -Fc fusion proteins or control.
  • Figures 75A-75B depict CD8+ T cell (CD8+CD45RA+) counts on Days (A) 7 and (B) 11 after treatment of NSG mice engrafted with human PBMCs with the indicated IL15/R -Fc fusion proteins or control.
  • Figures 76A-76B depict CD4+ T cell (CD4+CD45RA+) counts on Days A) 7 and B) 11 after treatment of NSG mice engrafted with human PBMCs with the indicated IL15/R -Fc fusion proteins or control.
  • Figure 77 depicts IFNy level on Days 4, 7, and 11 in serum of huPBMC engrafted mice following treatment with additional variant IL-15/R -Fc fusion proteins.
  • Figures 78A-78C depict CD8+ T cell count on Days (A) 4, (B) 7, and (C) 11 in whole blood of huPBMC engrafted mice following treatment with additional variant IL- 15/R -Fc fusion proteins.
  • Figures 79A-79C depict CD4+ T cell count on Days (A) 4, (B) 7, and (C) 11 in whole blood of huPBMC engrafted mice following treatment with additional variant IL- 15/R -Fc fusion proteins.
  • Figures 80A-80C depict CD45+ cell count on Days (A) 4, (B) 7, and (C) 11 in whole blood of huPBMC engrafted mice following treatment with additional variant IL- 15/R -Fc fusion proteins.
  • Figures 81A-81C depict the body weight as a percentage of initial body weight of huPBMC engrafted mice on Days (A) 4, (B) 7, and (C) 11 following treatment with additional IL-15/R variants. Each point represents a single NSG mouse. Mice whose body weights dropped below 70% initial body weight were euthanized. Dead mice are represented as 70%.
  • Figures 82A-82E depict lymphocyte counts after dosing cynomolgus monkeys with XENP20818.
  • Figures 82A-E respectively show the fold change in absolute count of CD56+ NK cells (Figure 82A), CD16+ NK cells ( Figure 82B), ⁇ T cells (CD45RA+CD3+CD4- CD8-) ( Figure 82C), CD8+ T cells ( Figure 82D), and CD4+ T cells ( Figure 82E).
  • Figures 83 A-83E depict proliferation of CD56+ NK cells ( Figure 83 A), CD16+ NK cells ( Figure 83B), CD8+ T cells (CD45RA+) ( Figure 83C), CD8+ T cells (CD45RA-) ( Figure 83D), and CD4+ T cells (CD45RA-) ( Figure 83E) after dosing cynomolgus monkeys with XENP20818.
  • Figures 84A-84E depict lymphocyte counts after dosing cynomolgus monkeys with XENP22819.
  • Figures 84A-E respectively show the fold change in absolute count of CD56+ NK cells (Figure 84A), CD16+ NK cells ( Figure 84B), ⁇ T cells (CD45RA+CD3+CD4- CD8-) ( Figure 84C), CD8+ T cells ( Figure 84D), and CD4+ T cells ( Figure 84E).
  • Figures 85 A-85E depict proliferation of CD56+ NK cells ( Figure 85 A), CD16+ NK cells ( Figure 85B), CD8+ T cells (CD45RA+) ( Figure 85C), CD8+ T cells (CD45RA-) ( Figure 85D), and CD4+ T cells (CD45RA-) ( Figure 85E) after dosing cynomolgus monkeys with XENP22819.
  • Figures 86A-86E depict lymphocyte counts after dosing cynomolgus monkeys with XENP22821.
  • Figures 86A-E respectively show the fold change in absolute count of CD56+ NK cells (Figure 86 A), CD16+ NK cells ( Figure 86B), ⁇ T cells (CD45RA+CD3+CD4- CD8-) ( Figure 86C), CD8+ T cells ( Figure 86D), and CD4+ T cells ( Figure 86E).
  • Figures 87A-87D depict proliferation of CD56+ NK cells (Figure 87A), CD16+ NK cells ( Figure 87B), CD8+ T cells (CD45RA+) ( Figure 87C), CD8+ T cells (CD45RA-) ( Figure 87D), and CD4+ T cells (CD45RA-) ( Figure 87E) after dosing cynomolgus monkeys with XENP22821.
  • Figures 88A-88E depict lymphocyte counts after dosing cynomolgus monkeys with XENP22822.
  • Figures 88A-E respectively show the fold change in absolute count of CD56+ NK cells (Figure 88 A), CD16+ NK cells ( Figure 88B), ⁇ T cells (CD45RA+CD3+CD4- CD8-) ( Figure 88C), CD8+ T cells ( Figure 88D), and CD4+ T cells ( Figure 88E).
  • Figures 89 A-89E depict proliferation of CD56+ NK cells (Figure 89 A), CD16+ NK cells ( Figure 89B), CD8+ T cells (CD45RA+) ( Figure 89C), CD8+ T cells (CD45RA-) ( Figure 89D), and CD4+ T cells (CD45RA-) ( Figure 89E) after dosing cynomolgus monkeys with XENP22822.
  • Figures 90A-90E depict lymphocyte counts after dosing cynomolgus monkeys with XENP22834.
  • Figures 90A-E respectively show the fold change in absolute count of CD56+ NK cells (Figure 90A), CD16+ NK cells ( Figure 90B), ⁇ T cells (CD45RA+CD3+CD4- CD8-) ( Figure 90C), CD8+ T ceUs ( Figure 90D), and CD4+ T cells ( Figure 90E).
  • Figures 91 A-91E depict proliferation of CD56+ NK cells (Figure 91 A), CD16+ NK cells ( Figure 91B), CD8+ T cells (CD45RA+) ( Figure 91C), CD8+ T cells (CD45RA-) ( Figure 91D), and CD4+ T cells (CD45RA-) ( Figure 91E) after dosing cynomolgus monkeys with XENP22834.
  • Figures 92A-92E depict lymphocyte counts after dosing cynomolgus monkeys with XENP23343.
  • Figures 92A-E respectively show the fold change in absolute count of CD56+ NK cells (Figure 92A), CD16+ NK cells ( Figure 92B), ⁇ T cells (CD45RA+CD3+CD4- CD8-) ( Figure 92C), CD8+ T cells ( Figure 92D), and CD4+ T cells ( Figure 92E).
  • Figures 93 A-93E depict proliferation of CD56+ NK cells (Figure 93 A), CD16+ NK cells ( Figure 93B), CD8+ T cells (CD45RA+) ( Figure 93C), CD8+ T cells (CD45RA-) ( Figure 93D), and CD4+ T cells (CD45RA-) ( Figure 93E) after dosing cynomolgus monkeys with XENP23343.
  • Figures 94A-94D depict sequences of XENP23343, XENP23504, XENP24113, XENP24301, XENP24306, and XENP24341, illustrative IL-15/R -Fc fusion proteins of the "IL-15/Ra-heteroFc" format with M428L/N434S substitutions.
  • IL-15 and IL-15R are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7), and slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 94D depicts sequences of XENP25938, an illustrative IL-15/R -Fc fusion protein of the "scIL- 15/R -Fc" format with M428L/N434S substitutions.
  • Figure 95 depicts sequences of XENP24383, an illustrative IL-15/R -Fc fusion protein of the "ncIL-15/Ra-Fc" format with M428L/N434S substitutions.
  • IL-15 and IL- 15R are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7)
  • slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figure 96 depicts sequences of XENP24346 and XENP24351, illustrative IL- 15/R -Fc fusion proteins of the "bivalent ncIL-15/R -Fc" format with M428L/N434S substitutions.
  • IL-15 and IL-15R are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in Figure 7), and slashes (/) indicate the border(s) between IL-15, IL-15R , linkers, and Fc regions.
  • Figures 97A-97C depict the percentage of Ki67 expression on (A) human CD8+ T cells, (B) human CD4+ T cells and (C) human NK cells following treatment with IL-15/R variants with M428L/N434S Fc mutations.
  • Figures 98A-98C depict the percentage of Ki67 expression on (A) cyno CD8+ T cells, (B) cyno CD4+ T cells and (C) cyno NK cells following treatment with IL-15/R variants with M428L/N434S Fc mutations.
  • Figures 99A-99C depict CD4+ T cell count on (A) Day 4 and (B) Day 7 in whole blood and (C) Day 8 in spleen of huPBMC engrafted mice following treatment with additional variant IL-15/R -Fc fusion proteins.
  • Figures lOOA-lOOC depict CD8+ T cell count on (A) Day 4 and (B) Day 7 in whole blood and (C) Day 8 in spleen of huPBMC engrafted mice following treatment with additional variant IL-15/R -Fc fusion proteins.
  • Figures 101A-101C depicts CD8+ T cell count on (A) Day 4 and (B) Day 7 in whole blood and (C) Day 8 in spleen of huPBMC engrafted mice following treatment with additional variant IL-15/R -Fc fusion proteins.
  • Figures 102A-102E depict the body weight as a percentage of initial body weight of huPBMC engrafted mice on Days (A) -2, (B) 1, (C) 5, (D) 8, and (E) 11 following treatment with additional IL-15/R variants. Each point represents a single NSG mouse.
  • Figure 102F depicts a time-course of body weight in huPBMC engrafted mice following treatment with the IL-15/R variants.
  • Figures 103A-103C depict (A) CD8+ T cell, (B) CD4+ T cell, and (C) NK cell counts in cynomolgus monkeys after treatment with IL-15/R variants on Day 1.
  • Figures 104A-104Z, 104AA-104AZ, and 104BA-104BL depicts sequences of the invention. The CDRs are in bold, IL-15 and IL15-R (sushi) are underlined, linkers are double underlined, and slashes (/) are between IL-15, IL15-R (sushi), linkers, and Fc domains.
  • Figure 105 depicts some preferred embodiments of the invention.
  • Xtend versions contain the 428L/434S variants in the Fc domains of each monomer.
  • Figure 106 depicts a list of engineered heterodimer-skewing (e.g. "steric heterodimerization") Fc variants with heterodimer yields (determined by HPLC-CIEX) and thermal stabilities (determined by DSC). Not determined thermal stability is denoted by "n.d.”.
  • engineered heterodimer-skewing e.g. "steric heterodimerization”
  • Fc variants with heterodimer yields determined by HPLC-CIEX
  • thermal stabilities determined by DSC. Not determined thermal stability is denoted by "n.d.”
  • ablation herein is meant a decrease or removal of activity.
  • “ablating FcyR binding” means the Fc region amino acid variant has less than 50% starting binding as compared to an Fc region not containing the specific variant, with less than 70-80-90-95-98% loss of activity being preferred, and in general, with the activity being below the level of detectable binding in a Biacore assay.
  • the Fc monomers of the invention retain binding to the FcRn receptor.
  • ADCC antibody dependent cell-mediated cytotoxicity
  • ADCC antibody dependent cell-mediated cytotoxicity
  • ADCC antibody dependent cell-mediated cytotoxicity
  • the cell-mediated reaction wherein nonspecific cytotoxic cells that express FcyRs recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • ADCC is correlated with binding to FcyRIIIa; increased binding to FcyRIIIa leads to an increase in ADCC activity.
  • ADCP or antibody dependent cell-mediated phagocytosis as used herein is meant the cell-mediated reaction wherein nonspecific cytotoxic cells that express FcyRs recognize bound antibody on a target cell and subsequently cause phagocytosis of the target cell.
  • modification herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence or an alteration to a moiety chemically linked to a protein.
  • a modification may be an altered carbohydrate or PEG structure attached to a protein.
  • amino acid modification herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence.
  • the amino acid modification is always to an amino acid coded for by DNA, e.g., the 20 amino acids that have codons in DNA and RNA.
  • amino acid substitution or “substitution” herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with a different amino acid.
  • the substitution is to an amino acid that is not naturally occurring at the particular position, either not naturally occurring within the organism or in any organism.
  • substitution E272Y refers to a variant polypeptide, in this case an Fc variant, in which the glutamic acid at position 272 is replaced with tyrosine.
  • a protein which has been engineered to change the nucleic acid coding sequence but not change the starting amino acid is not an ''amino acid substitution"; that is, despite the creation of a new gene encoding the same protein, if the protein has the same amino acid at the particular position that it started with, it is not an amino acid substitution.
  • amino acid insertion or “insertion” as used herein is meant the addition of an amino acid sequence at a particular position in a parent polypeptide sequence.
  • -233E or 233E designates an insertion of glutamic acid after position 233 and before position 234.
  • -233 ADE or A233ADE designates an insertion of AlaAspGlu after position 233 and before position 234.
  • amino acid deletion or “deletion” as used herein is meant the removal of an amino acid sequence at a particular position in a parent polypeptide sequence.
  • E233- or E233#, E233() or E233del designates a deletion of glutamic acid at position 233.
  • EDA233- or EDA233# designates a deletion of the sequence GluAspAla that begins at position 233.
  • variant protein or “protein variant”, or “variant” as used herein is meant a protein that differs from that of a parent protein by virtue of at least one amino acid modification.
  • Protein variant may refer to the protein itself, a composition comprising the protein, or the amino sequence that encodes it.
  • the protein variant has at least one amino acid modification compared to the parent protein, e.g. from about one to about seventy amino acid modifications, and preferably from about one to about five amino acid modifications compared to the parent.
  • the parent polypeptide for example an Fc parent polypeptide
  • the protein variant sequence herein will preferably possess at least about 80% identity with a parent protein sequence, and most preferably at least about 90% identity, more preferably at least about 95-98-99% identity .
  • Variant protein can refer to the variant protein itself, compositions comprising the protein variant, or the DNA sequence that encodes it.
  • antibody variant or “variant antibody” as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification
  • IgG variant or “variant IgG” as used herein is meant an antibody that differs from a parent IgG (again, in many cases, from a human IgG sequence) by virtue of at least one amino acid modification
  • immunoglobulin variant or “variant immunoglobulin” as used herein is meant an immunoglobulin sequence that differs from that of a parent immunoglobulin sequence by virtue of at least one amino acid modification.
  • Fc variant or “variant Fc” as used herein is meant a protein comprising an amino acid modification in an Fc domain.
  • the Fc variants of the present invention are defined according to the amino acid modifications that compose them.
  • N434S or 434S is an Fc variant with the substitution serine at position 434 relative to the parent Fc polypeptide, wherein the numbering is according to the EU index.
  • M428L/N434S defines an Fc variant with the substitutions M428L and N434S relative to the parent Fc polypeptide.
  • the identity of the WT amino acid may be unspecified, in which case the aforementioned variant is referred to as 428L/434S.
  • substitutions are provided is arbitrary, that is to say that, for example, 428L/434S is the same Fc variant as M428L/ 434S, and so on.
  • amino acid position numbering is according to the EU index.
  • the EU index or EU index as in Kabat or EU numbering scheme refers to the numbering of the EU antibody (Edelman et al., 1969, Proc Natl Acad Sci USA 63:78-85, hereby entirely incorporated by reference).
  • the modification can be an addition, deletion, or substitution.
  • substitutions can include naturally occurring amino acids and, in some cases, synthetic amino acids. Examples include U.S. Pat. No.
  • protein herein is meant at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides.
  • the peptidyl group may comprise naturally occurring amino acids and peptide bonds, or synthetic peptidomimetic structures, i.e. "analogs", such as peptoids (see Simon et al., PNAS USA 89(20):9367 (1992), entirely incorporated by reference).
  • the amino acids may either be naturally occurring or synthetic (e.g. not an amino acid that is coded for by DNA); as will be appreciated by those in the art.
  • homo-phenylalanine, dtrulline, ornithine and noreleucine are considered synthetic amino acids for the purposes of the invention, and both D- and L-(R or S) configured amino acids may be utilized.
  • the variants of the present invention may comprise modifications that include the use of synthetic amino acids incorporated using, for example, the technologies developed by Schultz and colleagues, including but not limited to methods described by Cropp & Shultz, 2004, Trends Genet.
  • polypeptides may include synthetic derivatization of one or more side chains or termini, glycosylation, PEGylation, circular permutation, cyclization, linkers to other molecules, fusion to proteins or protein domains, and addition of peptide tags or labels.
  • residue as used herein is meant a position in a protein and its associated amino acid identity.
  • Asparagine 297 also referred to as Asn297 or N297
  • Asn297 is a residue at position 297 in the human antibody IgGl.
  • IgG subclass modification or "isotype modification” as used herein is meant an amino acid modification that converts one amino acid of one IgG isotype to the corresponding amino acid in a different, aligned IgG isotype.
  • IgGl comprises a tyrosine and IgG2 a phenylalanine at EU position 296, a F296Y substitution in IgG2 is considered an IgG subclass modification.
  • non-naturally occurring modification as used herein is meant an amino acid modification that is not isotypic.
  • the substitution 434S in IgGl, IgG2, IgG3, or IgG4 (or hybrids thereof) is considered a non-naturally occurring modification.
  • amino acid and “amino acid identity” as used herein is meant one of the 20 naturally occurring amino acids that are coded for by DNA and RNA.
  • effector function as used herein is meant a biochemical event that results from the interaction of an antibody Fc region with an Fc receptor or ligand. Effector functions include but are not limited to ADCC, ADCP, and CDC.
  • IgG Fc ligand as used herein is meant a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an IgG antibody to form an Fc/Fc ligand complex.
  • Fc ligands include but are not limited to FcyRIs, FcyRIIs, FcyRIIIs, FcRn, Clq, C3, mannan binding lectin, mannose receptor, staphylococcal protein A, streptococcal protein G, and viral FcyR.
  • Fc ligands also include Fc receptor homologs (FcRH), which are a family of Fc receptors that are homologous to the FcyRs (Davis et al., 2002, Immunological Reviews 190:123-136, entirely incorporated by reference).
  • Fc ligands may include undiscovered molecules that bind Fc. Particular IgG Fc ligands are FcRn and Fc gamma receptors.
  • Fc ligand as used herein is meant a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an antibody to form an Fc/Fc ligand complex.
  • Fc gamma receptor any member of the family of proteins that bind the IgG antibody Fc region and is encoded by an FcyR gene.
  • this family includes but is not limited to FcyRI (CD64), including isoforms FcyRIa, FcyRIb, and FcyRIc; FcyRII (CD32), including isoforms FcyRIIa (including allotypes H131 and R131), FcyRIIb (including FcyRIIb-l and FcyRIIb-2), and FcyRIIc; and FcyRIII (CD16), including isoforms FcyRIIIa (including allotypes V158 and F158) and FcyRIIIb (including allotypes FcyRIIb-NAl and FcyRIIb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference), as well as any undiscovered human FcyRs or FcyR isoforms or allotypes.
  • An FcyR may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • Mouse FcyRs include but are not limited to FcyRI (CD64), FcyRII (CD32), FcyRIII (CD16), and FcyRIII-2 (CD16-2), as well as any undiscovered mouse FcyRs or FcyR isoforms or allotypes.
  • FcRn or "neonatal Fc Receptor” as used herein is meant a protein that binds the IgG antibody Fc region and is encoded at least in part by an FcRn gene.
  • the FcRn may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • the functional FcRn protein comprises two polypeptides, often referred to as the heavy chain and light chain.
  • the light chain is beta-2-microglobulin and the heavy chain is encoded by the FcRn gene.
  • FcRn or an FcRn protein refers to the complex of FcRn heavy chain with beta-2-microglobulin.
  • FcRn variants can be used to increase binding to the FcRn receptor, and in some cases, to increase serum half -life.
  • the Fc monomers of the invention retain binding to the FcRn receptor (and, as noted below, can include amino acid variants to increase binding to the FcRn receptor).
  • parent polypeptide as used herein is meant a starting polypeptide that is subsequently modified to generate a variant.
  • the parent polypeptide may be a naturally occurring polypeptide, or a variant or engineered version of a naturally occurring polypeptide.
  • Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it.
  • parent immunoglobulin as used herein is meant an unmodified immunoglobulin polypeptide that is modified to generate a variant
  • parent antibody as used herein is meant an unmodified antibody that is modified to generate a variant antibody. It should be noted that "parent antibody” includes known commercial, recombinantly produced antibodies as outlined below.
  • Fc or "Fc region” or “Fc domain” as used herein is meant the polypeptide comprising the constant region of an antibody excluding, in some instances, the first constant region immunoglobulin domain (e.g., CHI) or a portion thereof, and in some cases, part of the hinge.
  • an Fc can refer to the last two constant region immunoglobulin domains (e.g., CH2 and CH3) of IgA, IgD, and IgG, the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains.
  • Fc may include the J chain.
  • the Fc domain comprises immunoglobulin domains Cy2 and Cy3 (Cy2 and Cy3) and the lower hinge region between Cyl (Cyl) and Cy2 (Cy2).
  • an Fc refers to a truncated CHI domain, and CH2 and CH3 of an immunoglobulin.
  • the human IgG heavy chain Fc region is usually defined to include residues E216 or C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat.
  • amino acid amino acid
  • modifications are made to the Fc region, for example to alter binding to one or more FcyR receptors or to the FcRn receptor.
  • Fc fusion protein or “immunoadhesin” herein is meant a protein comprising an Fc region, generally linked (optionally through a linker moiety, as described herein) to a different protein, such as to IL-15 and/or IL-15R, as described herein.
  • two Fc fusion proteins can form a homodimeric Fc fusion protein or a
  • heterodimeric Fc fusion protein with the latter being preferred.
  • one monomer of the heterodimeric Fc fusion protein comprises an Fc domain alone (e.g., an empty Fc domain) and the other monomer is an Fc fusion, comprising a variant Fc domain and a protein domain, such as a receptor, ligand or other binding partner.
  • position as used herein is meant a location in the sequence of a protein. Positions may be numbered sequentially, or according to an established format, for example the EU index for antibody numbering.
  • strandedness in the context of the monomers of the heterodimeric antibodies of the invention herein is meant that, similar to the two strands of DNA that "match”, heterodimerization variants are incorporated into each monomer so as to preserve the ability to "match” to form heterodimers.
  • some pi variants are engineered into monomer A (e.g. making the pi higher) then steric variants that are "charge pairs” that can be utilized as well do not interfere with the pi variants, e.g. the charge variants that make a pi higher are put on the same "strand" or "monomer” to preserve both
  • wild type or WT herein is meant an amino acid sequence or a nucleotide sequence that is found in nature, including allelic variations.
  • a WT protein has an amino acid sequence or a nucleotide sequence that has not been intentionally modified.
  • the heterodimeric proteins of the present invention are generally isolated or recombinant.
  • isolated when used to describe the various polypeptides disclosed herein, means a polypeptide that has been identified and separated and/or recovered from a cell or cell culture from which it was expressed. Ordinarily, an isolated polypeptide will be prepared by at least one purification step.
  • isolated protein refers to aa protein which is substantially free of other antibodies having different antigenic specificities.
  • Recombinant means the proteins are generated using recombinant nucleic acid techniques in exogeneous host cells.
  • Percent (%) amino acid sequence identity with respect to a protein sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the specific (parental) sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. One particular program is the ALIGN-2 program outlined at paragraphs [0279] to [0280] of US Pub. No. 20160244525, hereby incorporated by reference.
  • invention sequence The degree of identity between an amino acid sequence of the present invention
  • parental amino acid sequence is calculated as the number of exact matches in an alignment of the two sequences, divided by the length of the "invention sequence,” or the length of the parental sequence, whichever is the shortest. The result is expressed in percent identity.
  • two or more amino acid sequences are at least 50%, 60%, 70%, 80%, or 90% identical. In some embodiments, two or more amino acid sequences are at least 95%, 97%, 98%, 99%, or even 100% identical.
  • Specific binding or “specifically binds to” or is "specific for” a particular antigen or an epitope means binding that is measurably different from a non-specific interaction. Specific binding can be measured, for example, by determining binding of a molecule compared to binding of a control molecule, which generally is a molecule of similar structure that does not have binding activity. For example, specific binding can be determined by competition with a control molecule that is similar to the target.
  • the present invention relates to heterodimeric Fc fusion proteins that include IL- 15 and IL-15 receptor alpha (IL-15R ) protein domains in different orientations.
  • the Fc domains can be derived from IgG Fc domains, e.g., IgGl, IgG2, IgG3 or IgG4 Fc domains, with IgGl Fc domains finding particular use in the invention.
  • each chain defines a constant region primarily responsible for effector function.
  • Kabat et al. collected numerous primary sequences of the variable regions of heavy chains and light chains. Based on the degree of conservation of the sequences, they classified individual primary sequences into the CDR and the framework and made a list thereof (see SEQUENCES OF IMMUNOLOGICAL INTEREST, 5th edition, NIH publication, No. 91-3242, E.A. Kabat et al., entirely incorporated by reference).
  • the Kabat numbering system is generally used when referring to a residue in the variable domain (approximately, residues 1-107 of the light chain variable region and residues 1-113 of the heavy chain variable region) and the EU numbering system for Fc regions (e.g., Kabat et al., supra (1991)).
  • immunoglobulin domains in the heavy chain.
  • immunoglobulin (Ig) domain herein is meant a region of an immunoglobulin having a distinct tertiary structure.
  • the heavy chain domains including, the constant heavy (CH) domains and the hinge domains.
  • the IgG isotypes each have three CH regions. Accordingly, "CH” domains in the context of IgG are as follows: “CHI” refers to positions 118-220 according to the EU index as in Kabat.
  • CH2 refers to positions 237-340 according to the EU index as in Kabat
  • CH3 refers to positions 341-447 according to the EU index as in Kabat.
  • the pi variants can be in one or more of the CH regions, as well as the hinge region, discussed below.
  • Ig domain of the heavy chain is the hinge region.
  • hinge region or “hinge region” or “antibody hinge region” or “immunoglobulin hinge region” herein is meant the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody.
  • the IgG CHI domain ends at EU position 220, and the IgG CH2 domain begins at residue EU position 237.
  • the antibody hinge is herein defined to include positions 221 (D221 in IgGl) to 236 (G236 in IgGl), wherein the numbering is according to the EU index as in Kabat.
  • the lower hinge is included, with the “lower hinge” generally referring to positions 226 or 230.
  • pi variants can be made in the hinge region as well.
  • the present invention provides different antibody domains.
  • the heterodimeric proteins of the invention comprise different domains, which can be overlapping as well. These domains include, but are not limited to, the Fc domain, the CHI domain, the CH2 domain, the CH3 domain, the hinge domain, and the heavy constant domain (CHl-hinge-Fc domain or CHl-hinge-CH2-CH3).
  • the "Fc domain” includes the -CH2-CH3 domain, and optionally a hinge domain.
  • the Fc domain also includes a truncated CHI domain.
  • a protein fragment e.g., IL-15 or IL-15R is attached to an Fc domain, it is the C-terminus of the IL-15 or IL-15R construct that is attached to all or part of the hinge of the Fc domain; for example, it is generally attached to the sequence EPKSS which is the beginning of the hinge.
  • a protein fragment e.g., IL-15 or IL-15R
  • it is the C-terminus of the IL-15 or IL15R construct that is attached to the CHI domain of the Fc domain.
  • the C-terminus of the IL-15 or IL-15R protein fragment is attached to the N-terminus of a domain linker, the C-terminus of which is attached to the N-terminus of a constant Fc domain (N-IL-15 or IL-15R protein fragment-linker-Fc domain-C) although that can be switched (N- Fc domain-linker- IL-15 or IL-15R protein fragment -C).
  • C-terminus of a first protein fragment is attached to the N- terminus of a second protein fragment, optionally via a domain linker
  • the C-terminus of the second protein fragment is attached to the N-terminus of a constant Fc domain, optionally via a domain linker.
  • a constant Fc domain that is not attached to a first protein fragment or a second protein fragment is provided.
  • a heterodimer Fc fusion protein can contain two or more of the exemplary monomeric Fc domain proteins described herein.
  • the linker is a "domain linker", used to link any two domains as outlined herein together, some of which are depicted in Figure 87. While any suitable linker can be used, many embodiments utilize a glycine-serine polymer, including for example (GS)n, (GSGGS)n, (GGGGS)n, and (GGGS)n, where n is an integer of at least one (and generally from 0 to 1 to 2 to 3 to 4 to 5) as well as any peptide sequence that allows for recombinant attachment of the two domains with sufficient length and flexibility to allow each domain to retain its biological function. In some cases, and with attention being paid to "strandedness", as outlined below, charged domain linkers.
  • GS glycine-serine polymer
  • heterodimeric Fc fusion proteins contain at least two constant domains which can be engineered to produce heterodimers, such as pi engineering.
  • Other Fc domains that can be used include fragments that contain one or more of the CHI, CH2, CH3, and hinge domains of the invention that have been pi engineered.
  • the formats depicted in Figures 9A-9G, and 39A-39D are heterodimeric Fc fusion proteins, meaning that the protein has two associated Fc sequences self -assembled into a
  • heterodimeric Fc domain and at least one protein fragment (e.g., 1, 2 or more protein fragments).
  • a first protein fragment is linked to a first Fc sequence and a second protein fragment is linked to a second Fc sequence.
  • a first protein fragment is linked to a first Fc sequence, and the first protein fragment is non-covalently attached to a second protein fragment that is not linked to an Fc sequence.
  • the heterodimeric Fc fusion protein contains a first protein fragment linked to a second protein fragment which is linked a first Fc sequence, and a second Fc sequence that is not linked to either the first or second protein fragments.
  • the present invention provides
  • heterodimeric Fc fusion proteins that rely on the use of two different heavy chain variant Fc sequences, that will self-assemble to form a heterodimeric Fc domain fusion polypeptide.
  • the present invention is directed to novel constructs to provide heterodimeric Fc fusion proteins that allow binding to one or more binding partners, ligands or receptors.
  • the heterodimeric Fc fusion constructs are based on the self-assembling nature of the two Fc domains of the heavy chains of antibodies, e.g., two "monomers” that assemble into a "dimer”.
  • Heterodimeric Fc fusions are made by altering the amino acid sequence of each monomer as more fully discussed below.
  • the present invention is generally directed to the creation of heterodimeric Fc fusion proteins which can co-engage binding partner(s) or ligand(s) or receptor(s) in several ways, relying on amino acid variants in the constant regions that are different on each chain to promote heterodimeric formation and/or allow for ease of purification of heterodimers over the homodimers.
  • heterodimers of the present invention are referred to as “heterodimerization variants”.
  • heterodimerization variants can include steric variants (e.g. the “knobs and holes” or “skew” variants described below and the “charge pairs” variants described below) as well as “pi variants", which allows purification of homodimers away from heterodimers.
  • heterodimerization variants useful mechanisms for heterodimerization include “knobs and holes” ("KIH”; sometimes herein as “skew” variants (see discussion in WO2014/145806), “electrostatic steering” or “charge pairs” as described in WO2014/145806, pi variants as described in WO2014/145806, and general additional Fc variants as outlined in WO2014/145806 and below.
  • KH knock-hole
  • skew electrostatic steering
  • charge pairs as described in WO2014/145806
  • pi variants as described in WO2014/145806
  • general additional Fc variants as outlined in WO2014/145806 and below.
  • pi variants can be either contained within the constant and/or Fc domains of a monomer, or domain linkers can be used. That is, the invention provides pi variants that are on one or both of the monomers, and/or charged domain linkers as well.
  • additional amino acid engineering for alternative functionalities may also confer pi changes, such as Fc, FcRn and KO variants.
  • amino acid variants can be introduced into one or both of the monomer polypeptides; that is, the pi of one of the monomers (referred to herein for simplicity as "monomer A”) can be engineered away from monomer B, or both monomer A and B change be changed, with the pi of monomer A increasing and the pi of monomer B decreasing.
  • the pi changes of either or both monomers can be done by removing or adding a charged residue (e.g., a neutral amino acid is replaced by a positively or negatively charged amino acid residue, e.g., glycine to glutamic acid), changing a charged residue from positive or negative to the opposite charge (e.g. aspartic acid to lysine) or changing a charged residue to a neutral residue (e.g., loss of a charge; lysine to serine.).
  • a charged residue e.g., a neutral amino acid is replaced by a positively or negatively charged amino acid residue, e.g., glycine to glutamic acid
  • changing a charged residue from positive or negative to the opposite charge e.g. aspartic acid to lysine
  • changing a charged residue to a neutral residue e.g., loss of a charge; lysine to serine.
  • this embodiment of the present invention provides for creating a sufficient change in pi in at least one of the monomers such that heterodimers can be separated from homodimers.
  • this can be done by using a "wild type" heavy chain constant region and a variant region that has been engineered to either increase or decrease its pi (wt A-+B or wt A - -B), or by increasing one region and decreasing the other region (A+ -B- or A- B+).
  • a component of some embodiments of the present invention are amino acid variants in the constant regions that are directed to altering the isoelectric point (pi) of at least one, if not both, of the monomers of a dimeric protein by incorporating amino acid substitutions ("pi variants" or "pi substitutions") into one or both of the monomers.
  • pi variants amino acid substitutions
  • pi substitutions amino acid substitutions
  • the separation of the heterodimers from the two homodimers can be accomplished if the pis of the two monomers differ by as little as 0.1 pH unit, with 0.2, 0.3, 0.4 and 0.5 or greater all finding use in the present invention.
  • the number of pi variants to be included on each or both monomer(s) to get good separation will depend in part on the starting pi of the components. As is known in the art, different Fes will have different starting pis which are exploited in the present invention. In general, as outlined herein, the pis are engineered to result in a total pi difference of each monomer of at least about 0.1 logs, with 0.2 to 0.5 being preferred as outlined herein.
  • the number of pi variants to be included on each or both monomer(s) to get good separation will depend in part on the starting pi of the components. That is, to determine which monomer to engineer or in which "direction" (e.g., more positive or more negative), the sequences of the Fc domains, and in some cases, the protein domain(s) linked to the Fc domain are calculated and a decision is made from there. As is known in the art, different Fc domains and/or protein domains will have different starting pis which are exploited in the present invention. In general, as outlined herein, the pis are engineered to result in a total pi difference of each monomer of at least about 0.1 logs, with 0.2 to 0.5 being preferred as outlined herein.
  • heterodimers can be separated from homodimers on the basis of size. As shown in the Figures, for example, several of the formats allow separation of heterodimers and homodimers on the basis of size.
  • heterodimerization variants including skew and purification heterodimerization variants
  • the possibility of immunogenicity resulting from the pi variants is significantly reduced by importing pi variants from different IgG isotypes such that pi is changed without introducing significant
  • an additional problem to be solved is the elucidation of low pi constant domains with high human sequence content, e.g. the minimization or avoidance of non-human residues at any particular position.
  • a side benefit that can occur with this pi engineering is also the extension of serum half-life and increased FcRn binding. That is, as described in USSN 13/194,904 (incorporated by reference in its entirety), lowering the pi of antibody constant domains (including those found in antibodies and Fc fusions) can lead to longer serum retention in vivo. These pi variants for increased serum half life also facilitate pi changes for purification.
  • the pi variants of the heterodimerization variants give an additional benefit for the analytics and quality control process of Fc fusion proteins, as the ability to either eliminate, minimize and distinguish when homodimers are present is significant. Similarly, the ability to reliably test the reproducibility of the heterodimeric Fc fusion protein production is important.
  • the present invention provides heterodimeric proteins, including heterodimeric Fc fusion proteins in a variety of formats, which utilize heterodimeric variants to allow for heterodimeric formation and/or purification away from homodimers.
  • the heterodimeric fusion constructs are based on the self-assembling nature of the two Fc domains, e.g., two "monomers” that assemble into a "dimer”.
  • these sets do not necessarily behave as "knobs in holes" variants, with a one-to-one correspondence between a residue on one monomer and a residue on the other; that is, these pairs of sets form an interface between the two monomers that encourages heterodimer formation and discourages homodimer formation, allowing the percentage of heterodimers that spontaneously form under biological conditions to be over 90%, rather than the expected 50% (25 % homodimer A/A:50% heterodimer A/B:25% homodimer B/B).
  • the formation of heterodimers can be facilitated by the addition of steric variants. That is, by changing amino acids in each heavy chain, different heavy chains are more likely to associate to form the heterodimeric structure than to form homodimers with the same Fc amino acid sequences.
  • Suitable steric variants are included in in the Figure 29 of USSN 15/141,350, all of which is hereby incorporated by reference in its entirety, as well as in Figure 84.
  • knocks and holes referring to amino acid engineering that creates steric influences to favor heterodimeric formation and disfavor homodimeric formation can also optionally be used; this is sometimes referred to as “knobs and holes”, as described in USSN 61/596,846, Ridgway et al., Protein Engineering 9(7):617 (1996); Atwell et al., J. Mol. Biol. 1997 270:26; US Patent No. 8,216,805, all of which are hereby incorporated by reference in their entirety.
  • the Figures identify a number of "monomer A - monomer B" pairs that rely on “knobs and holes”.
  • these "knobs and hole” mutations can be combined with disulfide bonds to skew formation to heterodimerization.
  • electrostatic steering As described in Gunasekaran et al., J. Biol. Chem. 285(25):19637 (2010), hereby incorporated by reference in its entirety. This is sometimes referred to herein as "charge pairs”.
  • electrostatics are used to skew the formation towards heterodimerization. As those in the art will appreciate, these may also have an effect on pi, and thus on purification, and thus could in some cases also be considered pi variants. However, as these were generated to force heterodimerization and were not used as purification tools, they are classified as "steric variants”.
  • D221E/P228E/L368E paired with D221R/P228R/K409R e.g., these are "monomer corresponding sets
  • steric variants outlined herein can be optionally and independently incorporated with any pi variant (or other variants such as Fc variants, FcRn variants, etc.) into one or both monomers, and can be independently and optionally included or excluded from the proteins of the invention.
  • the pair "S364K/E357Q : L368D/K370S” means that one of the monomers has the double variant set S364K E357Q and the other has the double variant set L368D/ 370S; as above, the "strandedness" of these pairs depends on the starting pi.
  • pi variants those that increase the pi of the protein (basic changes) and those that decrease the pi of the protein (acidic changes).
  • basic changes those that increase the pi of the protein
  • acidic changes those that decrease the pi of the protein
  • all combinations of these variants can be done: one monomer may be wild type, or a variant that does not display a significantly different pi from wild-type, and the other can be either more basic or more acidic. Alternatively, each monomer is changed, one to more basic and one to more acidic.
  • a preferred combination of pi variants has one monomer comprising 208D/295E/384D/418E/421D variants (N208D/Q295E/N384D/Q418E/ 421D when relative to human IgGl) if one of the Fc monomers includes a CHI domain.
  • the second monomer comprising a positively charged domain linker, including (GKPGS)4.
  • the first monomer includes a CHI domain, including position 208.
  • a preferred negative pi variant Fc set includes 295E/384D/418E/421D variants
  • mutations are made in the hinge domain of the Fc domain, including positions 221, 222, 223, 224, 225, 233, 234, 235 and 236. It should be noted that changes in 233-236 can be made to increase effector function (along with 327A) in the IgG2 backbone. Thus, pi mutations and particularly substitutions can be made in one or more of positions 221-225, with 1, 2, 3, 4 or 5 mutations finding use in the present invention. Again, all possible combinations are contemplated, alone or with other pi variants in other domains.
  • substitutions that find use in lowering the pi of hinge domains include, but are not limited to, a deletion at position 221, a non-native valine or threonine at position 222, a deletion at position 223, a non-native glutamic acid at position 224, a deletion at position 225, a deletion at position 235 and a deletion or a non-native alanine at position 236.
  • a deletion at position 221 a non-native valine or threonine at position 222
  • a deletion at position 223, a non-native glutamic acid at position 224 a deletion at position 225, a deletion at position 235 and a deletion or a non-native alanine at position 236.
  • substitution(s) are added to other pi variants in other domains in any combination.
  • mutations can be made in the CH2 region, including positions 274, 296, 300, 309, 320, 322, 326, 327, 334 and 339. Again, all possible combinations of these 10 positions can be made; e.g., a pi antibody may have 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 CH2 pi substitutions.
  • substitutions that find use in lowering the pi of CH2 domains include, but are not limited to, a non-native glutamine or glutamic acid at position 274, a non-native phenylalanine at position 296, a non native phenylalanine at position 300, a non-native valine at position 309, a non-native glutamic acid at position 320, a non-native glutamic acid at position 322, a non-native glutamic acid at position 326, a non-native glycine at position 327, a non-native glutamic acid at position 334, a non native threonine at position 339, and all possible combinations within CH2 and with other domains.
  • the mutations can be independently and optionally selected from position 355, 359, 362, 384, 389,392, 397, 418, 419, 444 and 447.
  • Specific substitutions that find use in lowering the pi of CH3 domains include, but are not limited to, a non native glutamine or glutamic acid at position 355, a non-native serine at position 384, a non-native asparagine or glutamic acid at position 392, a non-native methionine at position 397, a non native glutamic acid at position 419, a non native glutamic acid at position 359, a non native glutamic acid at position 362, a non native glutamic acid at position 389, a non native glutamic acid at position 418, a non native glutamic acid at position 444, and a deletion or non-native aspartic acid at position 447.
  • IgGl is a common isotype for therapeutic antibodies for a variety of reasons, including high effector function.
  • the heavy constant region of IgGl has a higher pi than that of IgG2 (8.10 versus 7.31).
  • IgG2 residues at particular positions into the IgGl backbone By introducing IgG2 residues at particular positions into the IgGl backbone, the pi of the resulting monomer is lowered (or increased) and additionally exhibits longer serum half-life.
  • IgGl has a glycine (pi 5.97) at position 137, and IgG2 has a glutamic acid (pi 3.22); importing the glutamic acid will affect the pi of the resulting protein.
  • a number of amino acid substitutions are generally required to significant affect the pi of the variant Fc fusion protein.
  • even changes in IgG2 molecules allow for increased serum half-life.
  • non-isotypic amino acid changes are made, either to reduce the overall charge state of the resulting protein (e.g., by changing a higher pi amino acid to a lower pi amino acid), or to allow accommodations in structure for stability, etc. as is more further described below.
  • pi engineering both the heavy and light constant domains significant changes in each monomer of the heterodimer can be seen. As discussed herein, having the pis of the two monomers differ by at least 0.5 can allow separation by ion exchange chromatography or isoelectric focusing, or other methods sensitive to isoelectric point.
  • the pi of each monomer can depend on the pi of the variant heavy chain constant domain and the pi of the total monomer, including the variant heavy chain constant domain and the fusion partner.
  • the change in pi is calculated on the basis of the variant heavy chain constant domain, using the chart in the Figure 19 of US Publ. App. No. 2014/0370013.
  • which monomer to engineer is generally decided by the inherent pi of each monomer.
  • the pi variant decreases the pi of the monomer, they can have the added benefit of improving serum retention in vivo.
  • Fc amino acid modification In addition to pi amino acid variants, there are a number of useful Fc amino acid modification that can be made for a variety of reasons, including, but not limited to, altering binding to one or more FcyR receptors, altered binding to FcRn receptors, etc.
  • the proteins of the invention can include amino acid modifications, including the heterodimerization variants outlined herein, which includes the pi variants and steric variants.
  • Each set of variants can be independently and optionally included or excluded from any particular heterodimeric protein.
  • Fc substitutions that can be made to alter binding to one or more of the FcyR receptors.
  • Substitutions that result in increased binding as well as decreased binding can be useful.
  • ADCC antibody dependent cell-mediated cytotoxicity; the cell-mediated reaction wherein nonspecific cytotoxic cells that express FcyRs recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • FcyRIIb an inhibitory receptor
  • Amino acid substitutions that find use in the present invention include those listed in USSNs 11/124,620 (particularly Figure 41), 11/174,287, 11/396,495, 11/538,406, all of which are expressly incorporated herein by reference in their entirety and specifically for the variants disclosed therein.
  • Particular variants that find use include, but are not limited to, 236 A, 239D, 239E, 332E, 332D, 239D/332E, 267D, 267E, 328F, 267E/328F, 236A/332E, 239D/332E/330Y, 239D, 332E/330L, 243 A, 243L, 264A, 264V and 299T.
  • amino acid substitutions that increase affinity for FcyRIIc can also be included in the Fc domain variants outlined herein.
  • the substitutions described in, for example, USSNs 11/124,620 and 14/578,305 are useful.
  • Fc substitutions that find use in increased binding to the FcRn receptor and increased serum half -life, as specifically disclosed in USSN 12/341,769, hereby incorporated by reference in its entirety, including, but not limited to, 434S, 434A, 428L, 308F, 2591, 428L/434S, 259I/308F, 436I/428L, 4361 or V/434S, 436V/428L and 259I/308F/428L.
  • FcyR ablation variants or “Fc knock out (FcKO or KG 1 )” variants.
  • FcyR ablation variants or “Fc knock out (FcKO or KG 1 )” variants.
  • FcKO or KG 1 Fey receptors
  • ablation variants are depicted in Figure 31 of USSN 15/141,350, all of which are herein incorporated by reference in its entirety, and each can be independently and optionally included or excluded, with preferred aspects utilizing ablation variants selected from the group consisting of G236R/L328R,
  • E233P/L234V/L235A/G236del/S239K E233P/L234V/L235A/G236del/S267K
  • heterodimerization variants including skew and/or pi variants
  • skew and/or pi variants can be optionally and independently combined in any way, as long as they retain their "strandedness" or "monomer partition”.
  • all of these variants can be combined into any of the heterodimerization formats.
  • any of the heterodimerization variants, skew and pi are also independently and optionally combined with Fc ablation variants, Fc variants, FcRn variants, as generally outlined herein.
  • a monomeric Fc domain can comprise a set of amino acid substitutions that includes C220S/S267K/L368D/K370S or C220S/S267K/S364K/E357Q.
  • heterodimeric Fc fusion proteins can comprise skew variants (e.g., a set of amino acid substitutions as shown in Figures 1 A-IC of USSN 15/141,350, all of which are herein incorporated by reference in its entirety ), with particularly useful skew variants being selected from the group consisting of S364K/E357Q : L368D/ 370S;
  • T366S/L368A/Y407V/Y349C T366W/S354C, optionally ablation variants, optionally charged domain linkers and the heavy chain comprises pi variants.
  • the Fc domain comprising an amino acid substitution selected from the group consisting of: 236R, 239D, 239E, 243L, M252Y, V259I, 267D, 267E, 298 A, V308F, 328F, 328R, 330L, 332D, 332E, M428L, N434A, N434S, 236R/328R, 239D/332E, M428L, 236R/328F, V259I/V308F, 267E/328F, M428L/N434S, Y436I/M428L, Y436V/M428L, Y436I/N434S, Y436V/N434S, 239D/332E/330L, M252Y/S254T/T256E, V259I/V308F/M428L, E233P/L234V/L235A/G
  • a particular combination of skew and pi variants that finds use in the present invention is T366S/L368A/Y407V : T366W (optionally including a bridging disulfide, T366S/L368A/Y407V Y349C : T366W/S354C) with one monomer comprises Q295E/N384D/Q418E/N481D and the other a positively charged domain linker.
  • the "knobs in holes" variants do not change pi, and thus can be used on either monomer.
  • IL-15 and IL15Ra Protein Domains [00218]
  • the present invention provides heterodimeric Fc fusion proteins containing
  • the IL-15 complex can take several forms. As stated above, the IL-15 protein on its own is less stable than when complexed with the IL-15R protein. As is known in the art, the IL-15R protein contains a "sushi domain", which is the shortest region of the receptor that retains IL-15 binding activity. Thus, while heterodimeric fusion proteins comprising the entire IL-15R protein can be made, preferred embodiments herein include complexes that just use the sushi domain, the sequence of which is shown in the figures.
  • the IL-15 complexes generally comprises the IL-15 protein and the sushi domain of IL IL-15R (unless otherwise noted that the full length sequence is used, "IL-15R ", “IL-15R (sushi)” and “sushi” are used interchangeably throughout).
  • This complex can be used in three different formats. As shown in Figures 9A, the IL-15 protein and the IL-15R (sushi) are not covalently attached, but rather are self-assembled through regular ligand-ligand interactions. As is more fully described herein, it can be either the IL- 15 domain or the sushi domain that is covalently linked to the Fc domain (generally using an optional domain linker).
  • each of the IL-15 and sushi domains can be engineered to contain a cysteine amino acid, that forms a disulfide bond to form the complex as is generally shown in Figures39A-39D, again, with either the IL-15 domain or the sushi domain being covalently attached (using an optional domain linker) to the Fc domain.
  • the human IL-15 protein has the amino acid sequence set forth in NCBI Ref. Seq. No. NP_000576.1 or SEQ ID NO:l. In some cases, the coding sequence of human IL-15 is set forth in NCBI Ref. Seq. No. NM_000585.
  • An exemplary IL-15 protein of the Fc fusion heterodimeric protein outlined herein can have the amino acid sequence of SEQ ID NO:2 or amino acids 49-162 of SEQ ID NO:l. In some embodiments, the IL-15 protein has at least 90%, e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to SEQ ID NO:2.
  • the IL-15 protein has the amino acid sequence set forth in SEQ ID NO:2 and the amino acid substitution N72D. In other embodiments, the IL-15 protein has the amino acid sequence of SEQ ID NO:2 and one or more amino acid substitutions selected from the group consisting of C42S, L45C, Q48C, V49C, L52C, E53C, E87C, and E89C. Optionally, the IL-15 protein also has an N72D substitution.
  • the IL-15 protein of the Fc fusion protein can have 1, 2, 3, 4, 5, 6, 7, 8 or 9 amino acid substitutions.
  • the amino acid substitution(s) may be isosteric substitutions at the IL-15:IL-2
  • the human IL-15 protein has one or more amino acid substitutions selected from the group consisting of N1D, N4D, D8N, D30N, D61N, E64Q, N65D, Q108E, and any combination thereof.
  • the IL-15 protein has the amino acid substitution Q108E. In some cases, the IL-15 protein has 1, 2, 3, 4, 5, 6, 7, 8, or more amino acid substitutions.
  • the IL-15 protein can have a N1D, N4D, D8N, D30N, D61N, E64Q, N65D, or Q108E substitution.
  • the amino acid substitution can include N1D/D61N, N1D/E64Q, N4D/D61N, N4D E64Q, D8N/D61N, D8N E64Q, D61N E64Q, E64Q/Q108E, N1D/N4D D8N,
  • the IL-15 protein has the amino acid substitutions D30N E64Q/N65D.
  • the human IL-15 receptor alpha (IL-15R ) protein has the amino acid sequence set forth in NCBI Ref . Seq. No. NP_002180.1 or SEQ ID NO:3. In some cases, the coding sequence of human IL-15R is set forth in NCBI Ref. Seq. No.
  • An exemplary the IL-15R protein of the Fc fusion heterodimeric protein outlined herein can comprise or consist of the sushi domain of SEQ ID NO:3 (e.g., amino acids 31-95 of SEQ ID NO:3), or in other words, the amino acid sequence of SEQ ID NO:4.
  • the IL-15R protein has the amino acid sequence of SEQ ID NO:4 and an amino acid insertion selected from the group consisting of D96, P97, A98, D96/P97, D96/C97, D96/P97/A98, D96/P97/C98, and D96/C97/A98, wherein the amino acid position is relative to full-length human IL-15R protein or SEQ ID NO:3.
  • amino acid(s) such as D (e.g., Asp), P (e.g., Pro), A (e.g., Ala), DP (e.g., Asp-Pro), DC (e.g., Asp-Cys), DP A (e.g., Asp-Pro-Ala), DPC (e.g., Asp-Pro-Cys), or DCA (e.g., Asp-Cys- Ala)
  • D e.g., Asp
  • P e.g., Pro
  • A e.g., Ala
  • DP e.g., Asp-Pro
  • DC e.g., Asp-Cys
  • DP A e.g., Asp-Pro-Ala
  • DPC e.g., Asp-Pro-Cys
  • DCA e.g., Asp-Cys- Ala
  • the IL-15R protein has the amino acid sequence of SEQ ID NO:4 and one or more amino acid substitutions selected from the group consisting of K34C, A37C, G38C, S40C, and L42C, wherein the amino acid position is relative to SEQ ID NO:4.
  • the IL-15R protein can have 1, 2, 3, 4, 5, 6, 7, 8 or more amino acid mutations (e.g., substitutions, insertions and/or deletions).
  • the IL-15 protein and IL-15R protein are attached together via a linker.
  • the proteins are not attached via a linker.
  • the IL-15 protein and IL-15R protein are noncovalently attached.
  • the IL-15 protein is attached to an Fc domain via a linker.
  • the IL-15 protein is attached to an Fc domain directly, such as without a linker.
  • the IL-15R protein is attached to an Fc domain via a linker.
  • the IL-15R protein is attached to an Fc domain directly.
  • a linker is not used to attach the IL-15 protein or IL-15R protein to an Fc domain.
  • the linker is a "domain linker", used to link any two domains as outlined herein together. While any suitable linker can be used, many embodiments utilize a glycine-serine polymer, including for example (GS)n, (GSGGS)n, (GGGGS)n, and (GGGS)n, where n is an integer of at least 0 (and generally from 0 to 1 to 2 to 3 to 4 to 5) as well as any peptide sequence that allows for recombinant attachment of the two domains with sufficient length and flexibility to allow each domain to retain its biological function. In certain cases, useful linkers include (GGGGS)o or (GGGGS)i or (GGGGS)2. In some cases, and with attention being paid to "strandedness", as outlined below, charged domain linkers can be used as discussed herein and shown in Figure 7.
  • the heteroditneric fusion proteins of the invention have two functional components: an IL-15/IL- 15R (sushi) component and an Fc component, both of which can take different forms as outlined herein and both of which can be combined with the other component in any configuration.
  • the first and the second Fc domains can have a set of amino acid
  • substitutions selected from the group consisting of a) S267K/L368D/ 370S :
  • the first and/or the second Fc domains have an additional set of amino acid substitutions comprising Q295E/N384D/Q418E/ 421D, according to EU numbering.
  • the first and/or the second Fc domains have an additional set of amino acid substitutions consisting of G236R/L328R, E233P/L234V L235A/G236del/S239K, E233P/L234V/L235A/G236del/S267K, E233P/L234V/L235A/G236del/S239K/A327G,
  • the first and/or second Fc domains have 428L/434S variants for half life extension.
  • the heterodimeric fusion protein comprises two monomers.
  • the first monomer comprises (from N-to C-terminus) IL-15- optional domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • the second monomer comprises the IL-15/R (sushi) domain-optional domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q : L368D/K370S.
  • a preferred embodiment utilizes the IL-15 variant Q108E.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q : L368D/K370S.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q : L368D/K370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants and the skew variant pair S364K/E357Q : L368D/ 370S.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q : L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N65D variant.
  • a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q : L368D/ 370S
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K E357Q : L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D/ 65D variant.
  • a preferred embodiment utilizes the IL-15 N4D/ 65D variant and the skew variant pair S364K/E357Q : L368D/ 370S.
  • a preferred embodiment utilizes the IL-15 N4D/ 65D variant, the skew variant pair S364K/E357Q : L368D/ 370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N1D/ 65D variant.
  • a preferred embodiment utilizes the IL-15 N1D/ 65D variant and the skew variant pair S364K/E357Q : L368D/ 370S.
  • a preferred embodiment utilizes the IL-15 N1D/ 65D variant, the skew variant pair S364K/E357Q : L368D/ 370S and the 428L/434S variants on each Fc monomer.
  • Figure 48 A XENP22822 including chain 1 (17693) and chain 2 (15908)
  • Figure 94 A XENP23504 including chain 1 and chain 2)
  • Figure 104 AO XENP24045 including chain 1 and chain 2)
  • Figure 104AQ XENP24306 including chain 1 and chain 2
  • Figure 48A XENP22821 including chain 1 and chain 2)
  • Figure 94A XENP23343 including chain 1 and chain 2)
  • Figure 104AJ XENP23557 including chain 1 and chain 2)
  • Figure 104AP ⁇ 24 ⁇ 3 including chain 1 and chain 2)
  • Figure 104AP XENP24051 including chain 1 and chain 2)
  • Figure 104AR XENP24341 including chain 1 and chain 2)
  • Figure 104AP XENP24052 including chain 1 and chain 2)
  • Figure 104AP XENP24301 including chain 1 and chain 2).
  • the heterodimeric fusion protein comprises two monomers.
  • the first monomer comprises (from N-to C-terminus) IL- 15/R (sushi)-domain linker-IL-15-optional domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • the second monomer comprises and "empty" Fc, comprising all or part of the hinge-CH2-CH3. This is referred to as "scIL- 15/R -Fc" with the "sc” standing for "single chain” (e.g. of the IL-15/sushi complex).
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q : L368D/ 370S.
  • a preferred embodiment utilizes the IL-15 variant Q108E.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K E357Q : L368D/ 370S.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K E357Q : L368D/ 370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15
  • a preferred embodiment utilizes the IL-15
  • a preferred embodiment utilizes the IL-15
  • a preferred embodiment utilizes the IL-15 N65D variant.
  • a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q : L368D/ 370S
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q : L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15
  • a preferred embodiment utilizes the IL-15
  • a preferred embodiment utilizes the IL-15
  • N4D/ 65D variant the skew variant pair S364K/E357Q : L368D/ 370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15
  • a preferred embodiment utilizes the IL-15
  • a preferred embodiment utilizes the IL-15 N1D/ 65D variant, the skew variant pair S364K/E357Q : L368D/ 370S and the 428L/434S variants on each Fc monomer.
  • the heterodimeric fusion protein comprises three monomers.
  • the first monomer comprises (from N-to C-terminus) IL- 15/R (sushi)-domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • the second monomer comprises and "empty” Fc, comprising all or part of the hinge-CH2-CH3.
  • the third monomer is IL-15. This is referred to as "ncIL-15/R -Fc" with the "nc" standing for "non-covalent”).
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q : L368D/ 370S.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 variant Q108E.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K E357Q : L368D/ 370S.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K E357Q : L368D/ 370S and the 428L/434S variants on both monomers.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15
  • D30N/E64Q/N65D variants the skew variant pair S364K/E357Q : L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N65D variant.
  • a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q : L368D/ 370S
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q : L368D/ 370S and the 428L/434S variants on each Fc monomer.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N4D/ 65D variant.
  • a preferred embodiment utilizes the IL-15 N4D/ 65D variant and the skew variant pair S364K/E357Q : L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D/ 65D variant, the skew variant pair S364K/E357Q : L368D/ 370S and the 428L/434S variants on each Fc monomer.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N1D/ 65D variant.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N1D/ 65D variant and the skew variant pair S364K/E357Q : L368D/K370S.
  • ncIL-15/R -heteroFc format a preferred embodiment utilizes the IL-15 N1D/ 65D variant, the skew variant pair S364K/E357Q : L368D/ 370S and the 428L/434S variants on each Fc monomer.
  • the heterodimeric fusion protein comprises four monomers.
  • the first and second monomers comprise (from N-to C- terminus) IL-15/R (sushi)-domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • the third and fourth monomers comprise IL-15. This is referred to as "bivalent ncIL-15/R -Fc" with the "nc" standing for "non-covalent”).
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q : L368D/ 370S.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 variant Q108E.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q : L368D/K370S.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q : L368D/K370S and the 428L/434S variants on both monomers.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants and the skew variant pair S364K/E357Q : L368D/ 370S.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q : L368D/K370S and the 428L/434S variants on each Fc monomer.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N65D variant.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q : L368D/ 370S
  • ncIL-15/R -Fc format In the bivalent ncIL-15/R -Fc format, a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K E357Q : L368D/K370S and the 428L/434S variants on each Fc monomer. [00294] In the bivalent ncIL-15/R -Fc format, a preferred embodiment utilizes the IL-15 N4D/ 65D variant.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N4D/ 65D variant and the skew variant pair S364K/E357Q : L368D/ 370S.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N4D/ 65D variant, the skew variant pair S364K/E357Q : L368D/ 370S and the 428L/434S variants on each Fc monomer.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N1D/ 65D variant.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N1D/ 65D variant and the skew variant pair S364K/E357Q : L368D/ 370S.
  • ncIL-15/R -Fc format a preferred embodiment utilizes the IL-15 N1D/ 65D variant, the skew variant pair S364K/E357Q : L368D/ 370S and the 428L/434S variants on each Fc monomer.
  • heterodimeric fusion proteins of the present invention can take on a wide variety of configurations, as are generally depicted in Figures 9A-9G and Figures 39A-39D.
  • the amino acid sequences of exemplary fusion proteins are provided in 8A-8E, 10, 11, 12A, 12B, 13-15, 40A, 40B, 41A, 41B, 42, 43, 48A-48D, 49A-49C, 50A, 50B, 51, 52, 53, and 94A-94D.
  • first fusion protein comprising an IL-15 protein domain covalently attached using a first domain linker to the N-terminus of a first Fc domain
  • second fusion protein comprising an IL-15R protein domain covalently attached using a second domain linker to the N-terminus of a second Fc domain
  • IL-15/R hetero Fc and "dsIL-15/R hetero Fc”
  • XENP20818 include, but are not limited to, XENP20818, XENP20819, XENP21471, XENP21472, XENP21473,
  • a useful format of a heterodimer Fc fusion protein comprises a fusion protein comprising a first protein domain covalently attached to the N-terminus of a second protein domain via a first domain linker that is covalently attached to the N-terminus of a first Fc domain via a second domain linker, and a second Fc domain (e.g., an empty Fc domain).
  • the first protein domain is an IL-15R protein domain and the second protein domain is an IL-15 protein domain.
  • An exemplary embodiment of this format (“scIL-15/ R - Fc”) includes, but is not limited to, XENP21478.
  • Yet another useful of a heterodimer Fc fusion protein outlined herein comprises a fusion protein comprising a first protein domain covalently attached to the N-terminus a first Fc domain via a domain linker, a second Fc domain (e.g., an empty Fc domain), and a second protein domain that is noncovalently attached to the first protein domain.
  • the first protein domain is an IL-15 protein domain and the second protein domain is an IL-15R protein domain.
  • ncIL-15/ R -Fc includes, but is not limited to, XENP21479, XENP22357, XENP22354, XENP22355, XENP22356, XENP22357, XENP22358, XENP22359, XENP22360, XENP22361, XENP22362, XENP22363, XENP22364, XENP22365, XENP22366, XENP22637, XENP24348, XENP24349, and XENP24383.
  • Another useful format of a heterodimer Fc fusion protein outlined herein comprises a first fusion protein comprising a first protein domain covalently attached to the N-terminus of said first Fc domain via a first domain linker, a second fusion protein comprising a second protein domain covalently attached to the N-terminus of said second Fc domain via a second domain linker, a third protein domain noncovalently attached to said first protein domain of said first fusion protein, and a fourth protein domain noncovalently attached to said second protein domain of said second fusion protein.
  • the first and second protein domains are IL-15 R protein domains
  • the third and fourth protein domains are IL-15 protein domains.
  • bivalent ncIL-15/ R -Fc or “bivalent dsIL-15/ R -Fc”
  • XENP21978 includes, but is not limited to, XENP21978, XENP22634, XENP24342, and XENP24346.
  • Yet another useful format of a heterodimer Fc fusion protein outlined herein comprises a first fusion protein comprising a first Fc domain covalently attached to the N- terminus of a first protein domain using a domain linker, a second Fc domain (e.g., an empty Fc domain), and a second protein domain noncovalently attached to said first protein domain.
  • An exemplary embodiment of this format (“Fc-ncIL-15/Ra" or "Fc-dsIL-15/Ra”) includes, but is not limited to, XENP22637 and XENP22639, and those depicted in Figure 16.
  • the first protein and the second protein are attached via a linker ( Figure 9G).
  • the first domain linker and the second domain linker can be the same or different.
  • the first Fc domain and the second Fc domain of the heterodimeric protein can have different amino acid sequences.
  • the Fc domains of the present invention comprise IgG Fc domains, e.g., IgGl Fc domains.
  • the first and second Fc domains comprising a set of amino acid substitutions selected from the group consisting of: L368D/ 370S and S364K;
  • L368D/ 370S and S364K/E357L L368D/K370S and S364K/E357Q; T411E/ 360E/Q362E and D401K; L368E/K370S and S364K; K370S and S364K/E357Q; K370S and S364K/E357Q;
  • the first and/or the second Fc domains of any of the heterodimeric Fc fusion formats outlined herein can have an additional set of amino acid substitutions comprising Q295E/ 384D/Q418E/N421D, according to EU numbering.
  • the first and/or the second Fc domains have an additional set of amino acid substitutions consisting of G236R/L328R, E233P/L234V/L235A/G236del/S239K, E233P L234V/L235A/G236del/S267K, E233P/L234V/L235A/G236del/S239K/A327G, E233P/L234V/L235A/G236del/S267K/A327G and E233P/L234V L235A/G236del, according to EU numbering.
  • compositions can further comprise ablation variants, pi variants, charged variants, isotypic variants, etc.
  • the invention further provides nucleic acid compositions encoding the heterodimeric Fc fusion protein of the invention (or, in the case of a monomer Fc domain protein, nucleic acids encoding those as well).
  • nucleic acid compositions will depend on the format of the heterodimeric protein.
  • the format requires three amino acid sequences
  • three nucleic acid sequences can be incorporated into one or more expression vectors for expression.
  • some formats only two nucleic acids are needed; again, they can be put into one or two expression vectors.
  • nucleic acids encoding the components of the invention can be incorporated into expression vectors as is known in the art, and depending on the host cells used to produce the heterodimeric Fc fusion proteins of the invention. Generally the nucleic acids are operably linked to any number of regulatory elements (promoters, origin of replication, selectable markers, ribosomal binding sites, inducers, etc.).
  • the expression vectors can be extra-chromosomal or integrating vectors.
  • nucleic acids encoding each monomer are each contained within a single expression vector, generally under different or the same promoter controls. In embodiments of particular use in the present invention, each of these two or three nucleic acids are contained on a different expression vector.
  • the heterodimeric Fc fusion protein of the invention are made by culturing host cells comprising the expression vector(s) as is well known in the art. Once produced, traditional fusion protein or antibody purification steps are done, including an ion exchange chromotography step. As discussed herein, having the pis of the two monomers differ by at least 0.5 can allow separation by ion exchange chromatography or isoelectric focusing, or other methods sensitive to isoelectric point.
  • the heterodimeric Fc fusion proteins of the invention are administered to patients with cancer, and efficacy is assessed, in a number of ways as described herein.
  • efficacy is assessed, in a number of ways as described herein.
  • standard assays of efficacy can be run, such as cancer load, size of tumor, evaluation of presence or extent of metastasis, etc.
  • immuno-oncology treatments can be assessed on the basis of immune status evaluations as well. This can be done in a number of ways, including both in vitro and in vivo assays. For example, evaluation of changes in immune status (e.g., presence of ICOS+ CD4+ T cells following ipi treatment) along with "old fashioned" measurements such as tumor burden, size, invasiveness, LN involvement, metastasis, etc.
  • any or all of the following can be evaluated: the inhibitory effects of PVRIG on CD4 + T cell activation or proliferation, CD8 + T (CTL) cell activation or proliferation, CD8 + T cell-mediated cytotoxic activity and/or CTL mediated cell depletion, NK cell activity and NK mediated cell depletion, the potentiating effects of PVRIG on Treg cell differentiation and proliferation and Treg- or myeloid derived suppressor cell (MDSC)- mediated immunosuppression or immune tolerance, and/or the effects of PVRIG on proinflammatory cytokine production by immune cells, e.g., IL-2, IFN-y or TNF- production by T or other immune cells.
  • CTL CD8 + T
  • CTL CTL cytotoxic activity and/or CTL mediated cell depletion
  • NK cell activity and NK mediated cell depletion the potentiating effects of PVRIG on Treg cell differentiation and proliferation and Treg- or myeloid derived suppressor cell (MDSC)- mediated immuno
  • assessment of treatment is done by evaluating immune cell proliferation, using for example, CFSE dilution method, Ki67 intracellular staining of immune effector cells, and 3 H-thymidine incorporation method,
  • assessment of treatment is done by evaluating the increase in gene expression or increased protein levels of activation-associated markers, including one or more of: CD25, CD69, CD137, ICOS, PD1, GITR, OX40, and cell degranulation measured by surface expression of CD107A.
  • assessment of treatment is done by assessing cytotoxic activity measured by target cell viability detection via estimating numerous cell parameters such as enzyme activity (including protease activity), cell membrane permeability, cell adherence, ATP production, co-enzyme production, and nucleotide uptake activity.
  • enzyme activity including protease activity
  • cell membrane permeability cell permeability
  • cell adherence cell adherence
  • ATP production co-enzyme production
  • nucleotide uptake activity include, but are not limited to, Trypan Blue or PI staining, 51 Cr or 35 S release method, LDH activity, MTT and/or WST assays, Calcein-AM assay, Luminescent based assay, and others.
  • assessment of treatment is done by assessing T cell activity measured by cytokine production, measure either intracellularly in culture supernatant using cytokines including, but not limited to, IFNy, TNF , GM-CSF, IL2, IL6, IL4, IL5, IL10, IL13 using well known techniques.
  • cytokines including, but not limited to, IFNy, TNF , GM-CSF, IL2, IL6, IL4, IL5, IL10, IL13 using well known techniques.
  • assessment of treatment can be done using assays that evaluate one or more of the following: (i) increases in immune response, (ii) increases in activation of ⁇ and/or ⁇ T cells, (iii) increases in cytotoxic T cell activity, (iv) increases in NK and/or NKT cell activity, (v) alleviation of ⁇ and/or ⁇ T-cell suppression, (vi) increases in pro- inflammatory cytokine secretion, (vii) increases in IL-2 secretion; (viii) increases in interferon-y production, (ix) increases in Thl response, (x) decreases in Th2 response, (xi) decreases or eliminates cell number and/or activity of at least one of regulatory T cells (Tregs).
  • T cell activation is assessed using a Mixed Lymphocyte Reaction (MLR) assay as is known in the art.
  • MLR Mixed Lymphocyte Reaction
  • the signaling pathway assay measures increases or decreases in immune response as measured for an example by phosphorylation or de- phosphorylation of different factors, or by measuring other post translational modifications.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in activation of ⁇ and/or ⁇ T cells as measured for an example by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in cytotoxic T cell activity as measured for an example by direct killing of target cells like for an example cancer cells or by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in NK and/or NKT cell activity as measured for an example by direct killing of target cells like for an example cancer cells or by cytokine secretion or by changes in expression of activation markers like for an example CD107a, etc. An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in ⁇ and/or ⁇ T-cell suppression, as measured for an example by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc. An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in pro-inflammatory cytokine secretion as measured for example by ELISA or by Luminex or by Multiplex bead based methods or by intracellular staining and FACS analysis or by Alispot etc. An increase in activity indicates immunostimulatory activity.
  • the signaling pathway assay measures increases or decreases in IL-2 secretion as measured for example by ELISA or by Luminex or by
  • the signaling pathway assay measures increases or decreases in interferon-y production as measured for example by ELISA or by Luminex or by Multiplex bead based methods or by intracellular staining and FACS analysis or by Alispot etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in Thl response as measured for an example by cytokine secretion or by changes in expression of activation markers.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in Th2 response as measured for an example by cytokine secretion or by changes in expression of activation markers. An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below. [00336] In one embodiment, the signaling pathway assay measures increases or decreases cell number and/or activity of at least one of regulatory T cells (Tregs), as measured for example by flow cytometry or by IHC. A decrease in response indicates immunostimulatory activity. Appropriate decreases are the same as for increases, outlined below.
  • the signaling pathway assay measures increases or decreases in M2 macrophages cell numbers, as measured for example by flow cytometry or by IHC. A decrease in response indicates immunostimulatory activity. Appropriate decreases are the same as for increases, outlined below.
  • the signaling pathway assay measures increases or decreases in M2 macrophage pro-tumorigenic activity, as measured for an example by cytokine secretion or by changes in expression of activation markers. A decrease in response indicates immunostimulatory activity. Appropriate decreases are the same as for increases, outlined below.
  • the signaling pathway assay measures increases or decreases in N2 neutrophils increase, as measured for example by flow cytometry or by IHC. A decrease in response indicates immunostimulatory activity. Appropriate decreases are the same as for increases, outlined below.
  • the signaling pathway assay measures increases or decreases in N2 neutrophils pro-tumorigenic activity, as measured for an example by cytokine secretion or by changes in expression of activation markers. A decrease in response indicates immunostimulatory activity. Appropriate decreases are the same as for increases, outlined below.
  • the signaling pathway assay measures increases or decreases in inhibition of T cell activation, as measured for an example by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc. An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in inhibition of CTL activation as measured for an example by direct killing of target cells like for an example cancer cells or by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc. An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in ⁇ and/or ⁇ T cell exhaustion as measured for an example by changes in expression of activation markers.
  • a decrease in response indicates immunostimulatory activity. Appropriate decreases are the same as for increases, outlined below.
  • the signaling pathway assay measures increases or decreases ⁇ and/or ⁇ T cell response as measured for an example by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in stimulation of antigen-specific memory responses as measured for an example by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD45RA, CCR7 etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below. .
  • the signaling pathway assay measures increases or decreases in apoptosis or lysis of cancer cells as measured for an example by cytotoxicity assays such as for an example MTT, Cr release, Calcine AM, or by flow cytometry based assays like for an example CFSE dilution or propidium iodide staining etc.
  • cytotoxicity assays such as for an example MTT, Cr release, Calcine AM
  • flow cytometry based assays like for an example CFSE dilution or propidium iodide staining etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in stimulation of cytotoxic or cytostatic effect on cancer cells, as measured for an example by cytotoxicity assays such as for an example MTT, Cr release, Calcine AM, or by flow cytometry based assays like for an example CFSE dilution or propidium iodide staining etc.
  • cytotoxicity assays such as for an example MTT, Cr release, Calcine AM
  • flow cytometry based assays like for an example CFSE dilution or propidium iodide staining etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases direct killing of cancer cells as measured for an example by cytotoxicity assays such as for an example MTT, Cr release, Calcine AM, or by flow cytometry based assays like for an example CFSE dilution or propidium iodide staining etc.
  • cytotoxicity assays such as for an example MTT, Cr release, Calcine AM
  • flow cytometry based assays like for an example CFSE dilution or propidium iodide staining etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases Thl7 activity as measured for an example by cytokine secretion or by proliferation or by changes in expression of activation markers.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in induction of complement dependent cytotoxicity and/or antibody dependent cell-mediated cytotoxicity, as measured for an example by cytotoxicity assays such as for an example MTT, Cr release, Calcine AM, or by flow cytometry based assays like for an example CFSE dilution or propidium iodide staining etc.
  • cytotoxicity assays such as for an example MTT, Cr release, Calcine AM, or by flow cytometry based assays like for an example CFSE dilution or propidium iodide staining etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • T cell activation is measured for an example by direct killing of target cells like for an example cancer cells or by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc.
  • increases in proliferation cell surface markers of activation (e.g., CD25, CD69, CD137, PD1), cytotoxicity (ability to kill target cells), and cytokine production (e.g., IL-2, IL- 4, IL-6, IFNy, TNF-a, IL-10, IL-17A) would be indicative of immune modulation that would be consistent with enhanced killing of cancer cells.
  • NK cell activation is measured for example by direct killing of target cells like for an example cancer cells or by cytokine secretion or by changes in expression of activation markers like for an example CD107a, etc.
  • increases in proliferation, cytotoxicity (ability to kill target cells and increases CD107a, granzyme, and perforin expression), cytokine production (e.g., IFNy and TNF ), and cell surface receptor expression (e.g. CD25) would be indicative of immune modulation that would be consistent with enhanced killing of cancer cells.
  • ⁇ T cell activation is measured for example by cytokine secretion or by proliferation or by changes in expression of activation markers.
  • Thl cell activation is measured for example by cytokine secretion or by changes in expression of activation markers.
  • compositions of the invention find use in a number of oncology applications, by treating cancer, generally by promoting T cell activation (e.g., T cells are no longer suppressed) with the binding of the heterodimeric Fc fusion proteins of the invention.
  • heterodimeric compositions of the invention find use in the treatment of these cancers.
  • Formulations of the antibodies used in accordance with the present invention are prepared for storage by mixing an antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (as generally outlined in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. [1980]), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, buffers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol;
  • buffers such as phosphate, citrate, and other organic acids
  • antioxidants including ascorbic acid and methionine
  • preservatives such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; al
  • resorcinol cyclohexanol; 3-pentanol; and m-cresol
  • low molecular weight polypeptides polypeptides
  • proteins such as serum albumin, gelatin, or immunoglobulins
  • hydrophilic polymers such as polyvinylpyrrolidone
  • amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine
  • chelating agents such as EDTA
  • sugars such as sucrose, mannitol, trehalose or sorbitol
  • salt-forming counter-ions such as sodium
  • metal complexes e.g. Zn-protein complexes
  • non-ionic surfactants such as TWEENTM, PLURONICSTM or polyethylene glycol (PEG).
  • heterodimeric proteins and chemotherapeutic agents of the invention are administered to a subject, in accord with known methods, such as intravenous
  • therapy is used to provide a positive therapeutic response with respect to a disease or condition.
  • positive therapeutic response is intended an improvement in the disease or condition, and/or an improvement in the symptoms associated with the disease or condition.
  • a positive therapeutic response would refer to one or more of the following improvements in the disease: (1) a reduction in the number of neoplastic cells; (2) an increase in neoplastic cell death; (3) inhibition of neoplastic cell survival; (5) inhibition (i.e., slowing to some extent, preferably halting) of tumor growth; (6) an increased patient survival rate; and (7) some relief from one or more symptoms associated with the disease or condition.
  • Positive therapeutic responses in any given disease or condition can be determined by standardized response criteria specific to that disease or condition.
  • Tumor response can be assessed for changes in tumor morphology (i.e., overall tumor burden, tumor size, and the like) using screening techniques such as magnetic resonance imaging (MRI) scan, x-radiographic imaging, computed tomographic (CT) scan, bone scan imaging, endoscopy, and tumor biopsy sampling including bone marrow aspiration (BMA) and counting of tumor cells in the circulation.
  • MRI magnetic resonance imaging
  • CT computed tomographic
  • BMA bone marrow aspiration
  • the subject undergoing therapy may experience the beneficial effect of an improvement in the symptoms associated with the disease.
  • Treatment according to the present invention includes a “therapeutically effective amount” of the medicaments used.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result.
  • a therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the medicaments to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects.
  • a "therapeutically effective amount" for tumor therapy may also be measured by its ability to stabilize the progression of disease.
  • the ability of a compound to inhibit cancer may be evaluated in an animal model system predictive of efficacy in human tumors.
  • this property of a composition may be evaluated by examining the ability of the compound to inhibit cell growth or to induce apoptosis by in vitro assays known to the skilled practitioner.
  • a therapeutically effective amount of a therapeutic compound may decrease tumor size, or otherwise ameliorate symptoms in a subject.
  • One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected.
  • Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • Parenteral compositions may be formulated in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • An exemplary, non imiting range for a therapeutically effective amount of an bispecific antibody used in the present invention is about 0.1-100 mg kg.
  • IL-15/IL-15R In order to address the short half-life of IL-15/IL-15R heterodimers, we generated the IL-15/IL-15R (sushi) complex as a Fc fusion (hereon referred to as IL-15/R -Fc fusion proteins) with the goal of facilitating production and promoting FcRn-mediated recycling of the complex and prolonging half-life.
  • IL-15/R -Fc fusion proteins herein referred to as IL-15/R -Fc fusion proteins
  • Example 1 A Engineering IL-15/R -Fc fusion proteins
  • Plasmids coding for IL-15 or IL-15R sushi domain were constructed by standard gene synthesis, followed by subcloning into a pTT5 expression vector containing Fc fusion partners (e.g., constant regions as depicted in Figures 8).
  • Cartoon schematics of illustrative IL-15/R -Fc fusion protein formats are depicted in Figures 9A-G.
  • the IL-15R heterodimeric Fc fusion or "IL-15/R -heteroFc" format comprises IL-15 recombinantly fused to one side of a heterodimeric Fc and IL-15R sushi domain recombinantly fused to the other side of the heterodimeric Fc ( Figure 9A).
  • the IL-15 and IL- 15R may have a variable length linker (see Figure 7) between their respective C-terminus and the N-terminus of the Fc region.
  • Illustrative proteins of this format include XENP20818 and XENP21475, sequences for which are depicted in Figure 10 (see also Table 1). Sequences for additional proteins of this format are listed as XENPs 20819, 21471, 21472, 21473, 21474, 21476, and 21477 in the figures and in the sequence listing.
  • the single-chain IL-15/R -Fc fusion or "scIL-15/R -Fc" format comprises IL- 15R sushi domain fused to IL-15 by a variable length linker (termed a "single-chain” IL- 15/IL-15R complex or "scIL-15/R ”) which is then fused to the N-terminus of a heterodimeric Fc-region, with the other side of the molecule being a "Fc-only” or "empty- Fc” heterodimeric Fc ( Figure 9B). Sequences for illustrative linkers are depicted in Figure 7.
  • XENP21478 An illustrative protein of this format is XENP21478, sequences for which are depicted in Figure 11 (also see Table 2). Sequences for additional proteins of this format are listed as XENPs 21993, 21994, 21995, 23174, 23175, 24477, and 24480 in the figures and the sequence listing.
  • the non-covalent IL-15/R -Fc fusion or "ncIL-15/R -Fc" format comprises IL- 15R sushi domain fused to a heterodimeric Fc region, while IL-15 is transfected separately so that a non-covalent IL-15/IL-15R complex is formed, with the other side of the molecule being a "Fc-only” or "empty-Fc" heterodimeric Fc ( Figure 9C).
  • Illustrative proteins of this format include XENP21479, XENP22366 and XENP24348, sequences for which are depicted in Figure 12.
  • the bivalent non-covalent IL-15/R -Fc fusion or "bivalent ncIL-15/R -Fc" format (Figure 9D) comprises IL-15R (sushi) fused to the N-terminus of a homodimeric Fc region, while IL-15 is transfected separately so that a non-covalent IL-15/R complex is formed.
  • An illustrative protein of this format is XENP21978, sequences for which are depicted in Figure 13. Sequences for additional proteins of this format are listed as XENP21979 in the figures and in the sequence listing.
  • the bivalent single-chain IL-15/R -Fc fusion or "bivalent scIL-15/R -Fc" format comprises IL-15 fused to IL-15R (sushi) by a variable length linker (termed a "single-chain” IL-15/IL-15R (sushi) complex or "scIL-15/R ”) which is then fused to the N- terminus of a homodimeric Fc-region. Sequences for illustrative linkers are depicted in Figure 7. Sequences for an illustrative protein of this format are depicted in Figure 14.
  • the Fc-non-covalent IL-15/R fusion or "Fc-ncIL-15/R " format comprises IL-15R (sushi) fused to the C-terminus of a heterodimeric Fc region, while IL-15 is transfected separately so that a non-covalent IL-15/R complex is formed, with the other side of the molecule being "Fc-only” or "empty Fc".
  • An illustrative protein of this format is XENP22637, sequences for which are depicted in Figure 15. Sequences for additional proteins of this format are listed XENP22638 in the figures and the sequence listing.
  • the Fc-single-chain IL-15/R fusion or "Fc-scIL-15/R " format comprises IL-15 fused to IL-15R (sushi) by a variable length linker ("scIL-15/R ") which is then fused to the C-terminus of a heterodimeric Fc region, with the other side of the molecule being "Fc-only” or "empty Fc". Sequences for illustrative linkers are depicted in Figure 7. Sequences for an illustrative protein of this format are depicted in Figure 16.
  • Proteins were produced by transient transfection in HEK293E cells and were purified by a two-step purification process comprising protein A chromatography (GE Healthcare) and anion exchange chromatography (HiTrapQ 5 mL column with a 5-40% gradient of 50 mM Tris pH 8.5 and 50 mM Tris pH 8.5 with 1 M NaCl).
  • Example IB Engineering IL-15/R -Fc fusion proteins
  • SEC size-exclusion chromatography
  • CEF capillary isoelectric focusing
  • the proteins were analyzed using SEC to measure their size (i.e. hydrodynamic volume) and determine the native-like behavior of the purified samples.
  • the analysis was performed on an Agilent 1200 high-performance liquid chromatography (HPLC) system. Samples were injected onto a SuperdexTM 200 10/300 GL column (GE Healthcare Life Sciences) at 1.0 mL/min using 1 x PBS, pH 7.4 as the mobile phase at 4oC for 25 minutes with UV detection wavelength at 280 nM. Analysis was performed using Agilent OpenLab Chromatography Data System (CDS) ChemStation Edition AIC version C.01.07.
  • CDS Agilent OpenLab Chromatography Data System
  • Example 1C Characterization of IL-15/R -Fc fusion proteins for affinity and stability
  • Affinity screens of IL-15/R -Fc fusion proteins were performed using Octet, a BioLayer Interferometry (BLI)-based method.
  • Experimental steps for Octet generally included the following: Immobilization (capture of ligand or test article onto a biosensor); Association (dipping of ligand- or test article-coated biosensors into wells containing serial dilutions of the corresponding test article or ligand); and Dissociation (returning of biosensors to well containing buffer) in order to determine the affinity of the test articles.
  • a reference well containing buffer alone was also included in the method for background correction during data processing.
  • anti-human Fc (AHC) biosensors were used to capture the test articles and then dipped into multiple concentration of IL-2R
  • the affinity results and corresponding sensorgrams are depicted in Figures 17D, 18D, and 19D.
  • Each of the three constructs showed high affinity binding (3-8 nM) for IL-lRp ⁇
  • IL-15/R -Fc fusion proteins in the various formats as described above were tested in a cell proliferation assay.
  • Human PBMCs were treated with the test articles at the indicated concentrations. 4 days after treatment, the PBMCs were stained with anti-CD8- FITC (RPA-T8), anti-CD4-PerCP/Cy5.5 (OKT4), anti-CD27-PE (M-T271), anti-CD56-BV421 (5.1H11), anti-CD16-BV421 (3G8), and anti-CD45RA-BV605 (HilOO) to gate for the foUowing cell types: CD4+ T cells, CD8+ T cells, and NK cells (CD56+/CD16+).
  • Ki67 is a protein strictly associated with cell proliferation, and staining for intracellular Ki67 was performed using anti-Ki67-APC (Ki-67) and Foxp3/Transcription Factor Staining Buffer Set (Thermo Fisher Scientific, Waltham, Mass.). The percentage of Ki67 on the above cell types was measured using FACS (depicted in Figures 20A-20C and 21A-21C).
  • the various IL-15/R -Fc fusion proteins induced strong proliferation of CD8+ T cells and NK cells. Notably, differences in proliferative activity were dependent on the linker length on the IL-15-Fc side. In particular, constructs having no linker (hinge only), including XENP21471, XENP21474, and XENP21475, demonstrated weaker proliferative activity.
  • Example IE Activity of IL-15/Ra-Fc fusion proteins in an SEB-stimulated PBMC assay
  • IL-15/R heterodimers can potently activate T cells.
  • IL- 15/R -Fc fusion proteins in the various formats as described above were tested in an SEB- stimulated PBMC assay.
  • Staphylococcal Enterotoxin B (SEB) is a superantigen that causes T cell activation and proliferation in a manner similar to that achieved by activation via the T cell receptor (TCR).
  • Stimulating human PBMC with SEB is a common method for assaying T cell activation and proliferation.
  • Human PBMCs from multiple donors were stimulated with 10 ng/mL of SEB for 72 hours in combination with 20 ⁇ g/mL of various IL-15/R -Fc fusion proteins or controls (PBS, anisotype control, and a bivalent anti-PD-1 antibody). After treatment, supernatant was collected and assayed for IL-2, data for which is depicted in Figure 22. The data clearly show that the IL-15/R -Fc fusion proteins enhanced IL-2 secretion more than PBS and isotype control. Notably, a number of the IL-15/R -Fc fusion proteins have activity equivalent to or better than that of the anti-PD-1 antibody.
  • Example IF IL-15/R -Fc fusion proteins enhance engraftment and disease activity in human PBMC -engrafted NSG mice
  • IL-15/R -Fc fusion protein XENP20818 was evaluated in a Graft-versus-Host Disease (GVHD) model conducted in female NSG (NOD-SCID-gamma) immunodeficient mice.
  • GVHD Graft-versus-Host Disease
  • NOD-SCID-gamma female NSG
  • IL-15/R -Fc fusion protein XENP20818 was evaluated in a Graft-versus-Host Disease (GVHD) model conducted in female NSG (NOD-SCID-gamma) immunodeficient mice.
  • GVHD Graft-versus-Host Disease
  • NOD-SCID-gamma NSG mice
  • Treatment of NSG mice injected with human PBMCs followed with IL-15/R -Fc fusion proteins enhances proliferation of the engrafted T cells.
  • Example 2 A Engineering and characterization of IL-15/R heterodimers with engineered disulfide bonds
  • Plasmids coding for IL-15 or IL-15R (sushi) were constructed by standard gene synthesis, followed by subcloning into a pTT5 expression vector.
  • the IL-15R (sushi) chain included a C-terminal polyhistidine tag. Residues identified as described above were substituted with cysteines by standard mutagenesis techniques. Additionally, up to three amino acids following the sushi domain in IL-15R were added to the C- terminus of IL- 15R (sushi) as a scaffold for engineering cysteines (illustrative sequences for which are depicted in Figure 27). Sequences for illustrative IL-15 and IL-15R (sushi) variants engineered with cysteines are respectively depicted in Figure 28 and 29.
  • FIG. 30A-C Cartoon schematics of IL-15/R heterodimers with and without engineered disulfides are depicted in Figures 30A-C. Sequences for an illustrative ncIL-15/R
  • heterodimer XENP21996 is depicted in Figure 31. Sequences for illustrative dsIL-15/R heterodimers XENP22004, XENP22005, XENP22006, XENP22008, and XENP22494 are depicted in Figure 32. Sequences for an illustrative scIL-15/R heterodimer are depicted in Figure 33. "Wild-type" IL-15/R heterodimers, with additional residues at the C-terminus but without engineered cysteines, were generated as controls. Sequences for these control IL-15/R heterodimers are listed as XENPs 22001, 22002, and 22003 in the figures and the sequence listing. Proteins were produced by transient transf ection in HEK293E cells and purified by Ni-NTA chromatography.
  • Example IB capillary isoelectric focusing
  • Example IB gel images for which are depicted in Figures 34-35.
  • the proteins were then screened for stability using DSF as generally described in Example 1C, data for which are depicted in Figures 36- 38.
  • the proteins were screened for binding to IL-2R
  • XENP22008 and XENP22494 and were constructed as Fc fusion proteins as described below.
  • Example 2B Characterization of IL-15/R -Fc fusion proteins with engineered disulfide bonds
  • Plasmids coding for IL-15 or IL-15R sushi domain with the above-described mutations were subdoned into a pTT5 expression vector containing Fc fusion partners (e.g., constant regions as depicted in Figures 8).
  • Cartoon schematics of IL-15/R -Fc fusion proteins with engineered disulfide bonds are depicted in Figures 39A-D.
  • Disulfide-bonded IL-15/R heterodimeric Fc fusion or "dsIL-15/R -heteroFc" (Figure 39A) is the same as "IL-15/R -heteroFc", but wherein IL-15R (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Illustrative proteins of this format include XENP22013, XENP22014, XENP22015, and XENP22017, sequences for which are depicted in Figure 40.
  • Disulfide-bonded IL-15/Ra Fc fusion or "dsIL-15/Ra-Fc" (Figure 39B) is the same as "ncIL-15/R -Fc", but wherein IL-15R (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Illustrative proteins of this format include XENP22357, XENP22358, XENP22359, XENP22684, and XENP22361, sequences for which are depicted in Figure 41. Sequences for additional proteins of this format are listed as XENPs 22360, 22362, 22363, 22364, 22365, and 22366 in the figures and the sequence listing.
  • Bivalent disulfide-bonded IL-15/R -Fc or "bivalent dsIL-15/R -Fc" (Figure 39C) is the same as "bivalent ncIL-15/R -Fc", but wherein IL-15R (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Illustrative proteins of this format include XENP22634, XENP22635, and XENP22636, sequences for which are depicted in Figure 42. Sequences for additional proteins of this format are listed as XENP22687 in the figures and the sequence listing.
  • Fc-disulfide-bonded IL-15/R fusion or "Fc-dsIL-15/R” ( Figure 39D) is the same as "Fc-ncIL-15/R ", but wherein IL-15R (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Illustrative proteins of this format include XENP22639 and XENP22640, sequences for which are depicted in Figure 43.
  • Proteins were produced by transient transfection in HEK293E cells and were purified by a two-step purification process comprising protein A chromatography (GE Healthcare) and anion exchange chromatography (HiTrapQ 5 mL column with a 5-40% gradient of 50 mM Tris pH 8.5 and 50 mM Tris pH 8.5 with 1 M NaCl).
  • IL-15/R -Fc fusion proteins (with or without engineered disulfide bonds) or controls were incubated with PBMCs for 4 days. Following incubation, PBMCs were stained with anti-CD4-PerCP/Cy5.5 (RPA-T4), anti-CD8-FITC (RPA-T8), anti-CD45RA-BV510 (HI100), anti-CD16-BV421 (3G8), anti-CD56-BV421 (HCD56), anti-CD27-PE (0323), and anti-Ki67-APC (Ki-67) to mark various cell populations and analyzed by FACS as generally described in Example ID.
  • NK cells Proliferation of NK cells, CD4+ T cells, and CD8+ T cells as indicated by Ki67 expression are depicted in Figures 45A-C.
  • Example 3 IL-15/R -Fc fusion proteins engineered for lower potency and increased PK and half-life
  • Example 3A Engineering and production of variant IL-15/R -Fc fusion
  • FIG. 46 depicts a structural model of the IL-15:receptor complexes showing locations of the predicted residues where we engineered isosteric substitutions (in order to reduce the risk of immunogenicity). Sequences for illustrative IL-15 variants engineered for reduced potency are depicted in Figure 47.
  • Plasmids coding for IL-15 or IL-15R were constructed by standard gene synthesis, followed by subcloning into a pTT5 expression vector containing Fc fusion partners (e.g., constant regions as depicted in Figures 8). Substitutions identified as described above were incorporated by standard mutagenesis techniques.
  • Proteins were produced by transient transfection in HEK293E cells and were purified by a two-step purification process comprising protein A chromatography (GE Healthcare) and anion exchange chromatography (HiTrapQ 5 mL column with a 5-40% gradient of 50 mM Tris pH 8.5 and 50 mM Tris pH 8.5 with 1 M NaCl).
  • Example 3B In vitro activity of variant IL-15/R -heteroFc and scIL-15/R -Fc fusion proteins engineered for decreased potency [00418] The variant IL-15/R -Fc fusion proteins were tested in a number of cell proliferation assays.
  • IL-15/R -Fc fusion proteins (with or without engineered substitutions) or control were incubated with PBMCs for 4 days. Following incubation, PBMCs were stained with anti-CD4-Evolve605 (SK-3), anti-CD8-PerCP/Cy5.5 (RPA-T8), anti-CD45RA-APC/Cy7 (HI100), anti-CD16-eFluor450 (CB16), anti-CD56- eFluor450 (TULY56), anti-CD3-FITC (OKT3), and anti-Ki67-APC (Ki-67) to mark various cell populations and analyzed by FACS as generally described in Example ID.
  • SK-3 anti-CD8-PerCP/Cy5.5
  • RPA-T8-PerCP/Cy5.5 anti-CD45RA-APC/Cy7
  • CB16 anti-CD16-eFluor450
  • TULY56 anti-CD3-FITC
  • Ki-67 anti-K
  • NK cells Proliferation of NK cells, CD8+ T cells, and CD4+ T cells as indicated by Ki67 expression are depicted in Figures 54-55. Most of the IL-15/R -Fc fusion proteins induced proliferation of each cell population; however, activity varied depending on the particular engineered substitutions.
  • IL-15/R -Fc fusion proteins (with or without engineered substitutions) were incubated with PBMCs for 3 days. Following incubation, PBMCs were stained with anti-CD3-FITC (OKT3), anti-CD4-Evolve604 (SK-3), anti-CD8- PerCP/Cy5.5 (RPA-T8), anti-CD16-eFluor450 (CB16), anti-CD56-eFluor450 (TULY56), anti- CD27-PE (0323), anti-CD45RA-APC/Cy7 (HI100) and anti-Ki67-APC (20Rajl) antibodies to mark various cell populations.
  • anti-CD3-FITC OKT3
  • SK-3 anti-CD4-Evolve604
  • RPA-T8- PerCP/Cy5.5 RPA-T8- PerCP/Cy5.5
  • CB16 anti-CD16-eFluor450
  • TULY56 anti-CD27-PE
  • Figures 56-57 depict selection of various cell populations following incubation with XENP22821 by FACS. Lymphocytes were first gated on the basis of side scatter (SSC) and forward scatter (FSC) (Figure 56A). Lymphocytes were then gated based on CD3 expression ( Figure 56B). Cells negative for CD3 expression were further gated based on CD16 expression to identify NK cells (CD16+) ( Figure 56C). CD3+ T cells were further gated based on CD4 and CD8 expression to identify CD4+ T cells, CD8+ T cells, and ⁇ T cells (CD3+CD4-CD8-) ( Figure 57 A). The CD4+ and CD8+ T cells were gated for CD45RA expression as shown respectively in Figures 57B-C.
  • SSC side scatter
  • FSC forward scatter
  • Figures 59A-D show the proliferation of the various cell populations. NK and CD8+ T cells are more sensitive than CD4+ T cells to IL-15/R -Fc fusion proteins, and as above, proliferative activity varied depending on the particular engineered substitutions.
  • Figure 59D shows the fold change in EC50 of various IL- 15/R -Fc fusion proteins relative to control XENP20818.
  • Figure 58A and B further depict the activation of lymphocytes following treatment with IL-15/R -Fc fusion proteins by gating for the expression of CD69 and CD25 (T cell activation markers) before and after incubation of PBMCs with XENP22821.
  • IL-15/R -Fc fusion proteins were incubated with human PBMCs for 3 days at 37°C. Following incubation, PBMCs were stained with anti-CD3-FITC (OKT3), anti-CD4-SB600 (SK-3), anti-CD8-PerCP/Cy5.5 (RPA- T8), anti-CD45RA-APC/Cy7 (HIlOO), anti-CD16-eFluor450 (CB16), anti-CD25-PE (M-A251), and anti-Ki67-APC (Ki-67) to mark various cell populations and analyzed by FACS as generally described in Example ID. Proliferation of CD8+ (CD45RA-) T cells, CD4+
  • PBMCs were incubated with the additional IL- 15/R -Fc variants at the indicated concentrations for 3 days. Following incubation, PBMCs were stained with anti-CD3-FITC (OKT3), anti-CD4 (SB600), anti-CD8-PerCP/Cy5.5 (RPA- T8), anti-CD16-eFluor450 (CB16), anti-CD25-PE (M-A251), anti-CD45RA-APC/Cy7 (HIlOO), and anti-Ki67-APC (Ki67) and analyzed by FACS as generally described in Example ID. Percentage of Ki67 on CD8+ T cells, CD4+ T cells and NK cells following treatment are depicted in Figure 61.
  • variant IL-15/R -Fc fusion proteins were incubated with human PBMCs for 3 days at 37°C. Following incubation, cells were stained with anti-CD3- PE (OKT3), anti-CD4-FITC (RPA-T4), anti-CD8 -BV510 (SKI), anti-CD8fi-APC (2ST8.5H7), anti-CD16-BV421 (3G8), anti-CD25-PerCP/Cy5.5 (M-A251), anti-CD45RA-APC/Cy7 (HIlOO), anti-CD56-BV605 (NCAM16.2), and anti-Ki67-PE/Cy7 (Ki-67) and analyzed by FACS as generally described in Example ID. Percentage of Ki67 on CD8+ T cells, CD4+ T cells, ⁇ T cells, and NK cells are depicted in Figures 62A-E.
  • variant IL-15/R -Fc fusion proteins were incubated with human PBMCs for 3 days at 37°C. Following incubation, cells were stained with anti-CD3- PE (OKT3), anti-CD4-FITC (RPA-T4), anti-CD8 -BV510 (SKI), anti-CD8fi-APC (SIDI8BEE), anti-CD16-BV421 (3G8), anti-CD25-PerCP/Cy5.5 (M-A251), anti-CD45RA-APC/Cy7 (HIlOO), anti-CD56-BV605 (NCAM16.2), and anti-Ki67-PE/Cy7 (Ki-67) and analyzed by FACS as generally described in Example ID. Percentage of Ki67 on CD8+ T cells, CD4+ T cells, ⁇ T cells, and NK cells are depicted in Figures 63A-E.
  • Example 3C In vitro activity of variant scIL-15/Ra-Fc fusion proteins
  • IL-15/R -Fc fusion proteins with some of the substitutions described above, further with different lengths linkers between IL-15 and IL-15R (as depicted in Table 3) were incubated with human PBMCs at the indicated concentrations for 3 days at 37°C.
  • PBMCs were stained with anti-CD3-PE (OKT3), anti-CD4-FITC (RPA- T4), anti-CD8-APC (RPA-T8), anti-CD16-BV605 (3G8), anti-CD25-PerCP/Cy5.5 (M-A251), anti-CD45RA-APC/Fire750 (HI100) and anti-Ki67-PE/Cy7 (Ki-67) and analyzed by FACS as generally described in Example ID. Percentage Ki67 on CD8+ T cells, CD4+ T cells, ⁇ T cells and NK (CD16+) cells are depicted in Figures 64A-D.
  • ncIL-15/R -Fc fusion protein XENP21479 is the most potent inducer of CD8+ T cell, CD4+ T cell, NK (CD16+) cell, and ⁇ T cell proliferation.
  • scIL-15/R -Fc fusion proteins were less potent than XENP21479 in inducing proliferation, but differences were dependent on both the linker length, as well as the particular engineered substitutions.
  • Example 3D In vitro activity of variant IL-15/R -Fc fusion proteins
  • Variant IL-15/R -Fc fusion proteins in different formats were incubated with human PBMCs at the indicated concentrations for 3 days at 37°C. Following incubation, PBMCs were stained with anti-CD3-PE (OKT3), anti-CD4-FITC (RPA- T4), anti-CD8-APC (RPA-T8), anti-CD16-BV605 (3G8), anti-CD25-PerCP/Cy5.5 (M-A251), anti-CD45RA-APC/Fire750 (HI100) and anti-Ki67-PE/Cy7 (Ki-67) and analyzed by FACS as generally described in Example ID.
  • ncIL-15/R -Fc fusion protein XENP21479 is the most potent inducer of CD8+ T cell, CD4+ T cell, NK (CD16+) cell, and ⁇ T cell proliferation.
  • introduction of Q108E substitution into the ncIL-15/R -Fc format drastically reduces its proliferative activity in comparison to wildtype (XENP21479).
  • Example 3E STAT5 phosphorylation by variant IL-15/Ra-Fc fusion proteins
  • Transpresentation of IL-15 and IL-15R drives phosphorylation of STAT5 and subsequent proliferation of NK and T cells (CD4+ and CD8+). Accordingly, CD8+ and CD4+ T cells were analyzed for STAT5 phosphorylation following 15 minutes incubation with the indicated IL-15/R -Fc test articles.
  • PBMCs were stained with anti-CD4-BV421 (RPA-T4) and anti-CD8-A700 (SKI) for 30-45 minutes at room temperature. Cells were washed and incubated with pre-chilled (-20°C) 90% methanol for 20-60 minutes.
  • FIGs 66A-D depict selection of various cell populations following incubation with XENP22821. Lymphocytes were first gated on the basis of SSC and FSC ( Figure 66 A). The lymphocytes were then gated based on CD4 and CD8 expression to identify CD4+ and CD8+ T cells ( Figure 66B).
  • Example 3F PK of variant IL-15/Ra-Fc fusion proteins engineered for lower potency
  • Example 3G Variant IL-15/R -Fc fusion proteins enhance engraftment and disease activity in human PBMC -engrafted NSG mice
  • variant IL-15/R -Fc fusion proteins were evaluated in a GVHD models conducted in female NSG immunodeficient mice as generally described in Example IF.
  • Example 3H IL-15/R -Fc fusion proteins are active in cynomolgus
  • Lymphocyte counts Figures 82, 84, 86, 88, 90, and 92
  • proliferation Figures 83, 85, 87, 89, 91, and 93 were assessed over time.
  • Figures show significant expression of Ki67 on CD56+ NK cells (Figure 87 A), CD16+ NK cells (Figure 87B), CD8+ T cells (CD45RA+) ( Figure 87C), CD8+ T cells (CD45RA-) ( Figure 87D), and CD4+ T cells (Figure 87E) indicating proliferative activity following treatment with XENP22821. Similar proliferative activity was observed following treatment with XENP20818, XENP22819, XENP22822, and XENP23343, demonstrating that most of the IL-15/R -Fc fusion proteins of the invention are active in cynomolgus monkeys.
  • IL-15/R -Fc variants engineered for decreased potency as described above were further engineered with Xtend Fc (hereon referred to as "IL-15/R -XtendFc" fusion proteins) to further increase half-life by subcloning plasmids coding for IL-15 and/or IL-15R (sushi) into a pTT5 expression vector containing Fc fusion partners with M428L/N434S substitutions (see Figure 8, Backbone 11). Sequences for illustrative IL-15/R -XtendFc are depicted in Figures 94-96 (see also Table 5).
  • Example 4A In vitro activity of additional IL-15/R -Fc variants
  • Human PBMCs were incubated with the IL-15/R -XtendFc variants at the indicated concentrations for 3 days. Following incubation, PBMCs were stained with anti- CD3-FITC (OKT3), anti-CD4-PE (RPA-T4), anti-CD8-eFluor450 (SK-1), anti-CD45RA-PE/Cy7 (HI100), anti-CD16-PerCP/Cy5.5 (3G8), anti-CD25-APC/Fire750 (M-A251), and anti-Ki67- APC (Ki-67) to mark various cell populations and analyzed by FACS as generally described in Example ID.
  • Proliferation of CD8+ T cells, CD4+ T cells and NK cells following treatment as indicated by Ki67 expression are depicted in Figure 97.
  • Xtend variants were selected for investigating activity in cynomolgus monkeys, their ability to proliferate cynomolgus T cells was investigated. Cyno PBMCs were incubated with selected test articles at the indicated concentrations for 3 days.
  • PBMCs were stained with anti-CD3-FITC (SP34), anti-CD4-PE/Cy7 (OKT4), anti- CD8-APC (RPA-T8), anti-CD45RA-APC/Fire750 (HI100), anti-CD16-BV605 (3G8), anti-CD25- BV421 (M-A251), and anti-Ki67-PerCP/Cy5.5 (Ki-67) to mark various cell populations and analyzed by FACS as generally described in Example ID. Proliferation of CD8+ T cells, CD4+ T cells and NK cells following treatment as indicated by Ki67 expression are depicted in Figure 98.
  • Example 4B In vivo activity of IL-15/Ra-XtendFc variants in a GVHD model
  • Figures 101A-C respectively depict CD4+ T cell counts on Days 4 and 7 in whole blood and Day 8 in spleen. Body weight of the mice were also measured on Day -8, -2, 1, 5, 8 and 11 as depicted in Figures 102A-102F. Each point represents one female NSG mouse.
  • Example 4C In vivo activity of variant IL-15/R -XtendFc fusion proteins in cynomolgus monkeys
  • CD8+ T cell, CD4+ T cell and NK cell counts in blood were assessed over time as depicted respectively in Figures 103A-C.
  • Each point is an average of 3 cynomolgus monkeys. The data show that each of the variants were active in proliferating immune cells indicating that the IL-15/R -Fc fusion proteins of the invention could be useful as therapeutics for cancer in humans.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
PCT/US2017/056829 2016-10-14 2017-10-16 IL15/IL15Rα HETERODIMERIC FC-FUSION PROTEINS WO2018071919A1 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
KR1020247008957A KR20240038176A (ko) 2016-10-14 2017-10-16 IL15/IL15Rα 이종이량체 Fc-융합 단백질
BR112019007281A BR112019007281A2 (pt) 2016-10-14 2017-10-16 proteína heterodimérica, composições de ácido nucleico e de vetor de expressão, vetor de expressão, célula hospedeira, e, métodos para produzir proteína heterodimérica e para tratar câncer em um paciente
PE2019000823A PE20191033A1 (es) 2016-10-14 2017-10-16 PROTEINAS DE FUSION FC HETERODIMERICAS IL 15/IL 15R(alfa)
SG11201903304YA SG11201903304YA (en) 2016-10-14 2017-10-16 IL15/IL15Ra HETERODIMERIC FC-FUSION PROTEINS
MX2019004328A MX2019004328A (es) 2016-10-14 2017-10-16 Proteínas de fusión fc heterodiméricas il15/il15ra.
CA3040504A CA3040504A1 (en) 2016-10-14 2017-10-16 Il15/il15ra heterodimeric fc-fusion proteins
CN201780067629.XA CN110214147A (zh) 2016-10-14 2017-10-16 IL15/IL15Rα异源二聚体FC-融合蛋白
RU2019114268A RU2779938C2 (ru) 2016-10-14 2017-10-16 ГЕТЕРОДИМЕРНЫЕ Fc-СЛИТЫЕ БЕЛКИ IL15/IL15Rα
CR20230179A CR20230179A (es) 2016-10-14 2017-10-16 PROTEÍNAS DE FUSIÓN FC HETERODIMÉRICAS IL 15/IL15Ra (Divisional 2019-0229)
AU2017342560A AU2017342560B2 (en) 2016-10-14 2017-10-16 IL15/IL15Ralpha heterodimeric Fc-fusion proteins
CR20190229A CR20190229A (es) 2016-10-14 2017-10-16 PROTEÍNAS DE FUSIÓN FC HETERODIMÉRICAS IL15/IL15Ra
EP17804322.0A EP3526241A1 (en) 2016-10-14 2017-10-16 Il15/il15r heterodimeric fc-fusion proteins
KR1020197012663A KR102649972B1 (ko) 2016-10-14 2017-10-16 IL15/IL15Rα 이종이량체 Fc-융합 단백질
JP2019519632A JP7142630B2 (ja) 2016-10-14 2017-10-16 IL15/IL15Rαヘテロ二量体FC-融合タンパク質
IL265998A IL265998B1 (en) 2016-10-14 2019-04-11 il15/il15rα heterodimeric fc-cocci proteins
PH12019500781A PH12019500781A1 (en) 2016-10-14 2019-04-11 IL15/IL15Ra HETERODIMERIC FC-FUSION PROTEINS
ZA2019/02380A ZA201902380B (en) 2016-10-14 2019-04-15 Il15/il15ralpha heterodimeric fc-fusion proteins
CONC2019/0003943A CO2019003943A2 (es) 2016-10-14 2019-04-17 Proteínas de fusión fc heterodiméricas il15/il15rα
AU2022203782A AU2022203782B2 (en) 2016-10-14 2022-06-01 IL15/IL15Ralpha heterodimeric Fc-fusion proteins
JP2022145394A JP2022180450A (ja) 2016-10-14 2022-09-13 IL15/IL15Rαヘテロ二量体FC-融合タンパク質
AU2024203151A AU2024203151A1 (en) 2016-10-14 2024-05-13 IL15/IL15Ralpha heterodimeric Fc-fusion proteins
IL312989A IL312989A (en) 2016-10-14 2024-05-20 il15/il15rα heterodimeric fc-cocci proteins

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201662408655P 2016-10-14 2016-10-14
US62/408,655 2016-10-14
US201662416087P 2016-11-01 2016-11-01
US62/416,087 2016-11-01
US201762443465P 2017-01-06 2017-01-06
US62/443,465 2017-01-06
US201762477926P 2017-03-28 2017-03-28
US62/477,926 2017-03-28

Publications (1)

Publication Number Publication Date
WO2018071919A1 true WO2018071919A1 (en) 2018-04-19

Family

ID=60452718

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2017/056829 WO2018071919A1 (en) 2016-10-14 2017-10-16 IL15/IL15Rα HETERODIMERIC FC-FUSION PROTEINS
PCT/US2017/056826 WO2018071918A1 (en) 2016-10-14 2017-10-16 Bispecific heterodimeric fusion proteins containing il-15/il-15ralpha fc-fusion proteins and pd-1 antibody fragments

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2017/056826 WO2018071918A1 (en) 2016-10-14 2017-10-16 Bispecific heterodimeric fusion proteins containing il-15/il-15ralpha fc-fusion proteins and pd-1 antibody fragments

Country Status (21)

Country Link
US (6) US10550185B2 (es)
EP (2) EP3526241A1 (es)
JP (4) JP7273453B2 (es)
KR (3) KR102649972B1 (es)
CN (2) CN110214148A (es)
AU (5) AU2017342560B2 (es)
BR (2) BR112019007288A2 (es)
CA (2) CA3040504A1 (es)
CL (4) CL2019001000A1 (es)
CO (2) CO2019003943A2 (es)
CR (2) CR20230179A (es)
IL (3) IL265997A (es)
MA (2) MA46533A (es)
MX (3) MX2019004328A (es)
PE (2) PE20191034A1 (es)
PH (2) PH12019500782A1 (es)
RU (1) RU2019114175A (es)
SA (1) SA519401562B1 (es)
SG (2) SG11201903304YA (es)
WO (2) WO2018071919A1 (es)
ZA (2) ZA201902383B (es)

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019006472A1 (en) * 2017-06-30 2019-01-03 Xencor, Inc. TARGETED HETETRODIMERIC FUSION PROTEINS CONTAINING IL-15 / IL-15RA AND ANTIGEN-BINDING DOMAINS
WO2019204646A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Lag-3 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and lag-3 antigen binding domains
WO2019204592A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof
WO2019204655A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Tim-3 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and tim-3 antigen binding domains
WO2019204665A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Pd-1 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and pd-1 antigen binding domains and uses thereof
WO2019229658A1 (en) 2018-05-30 2019-12-05 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-fusion proteins
WO2020086758A1 (en) * 2018-10-23 2020-04-30 Dragonfly Therapeutics, Inc. Heterodimeric fc-fused proteins
WO2020132646A1 (en) * 2018-12-20 2020-06-25 Xencor, Inc. Targeted heterodimeric fc fusion proteins containing il-15/il-15ra and nkg2d antigen binding domains
US10696722B2 (en) 2016-08-10 2020-06-30 Ajou University Industry-Academic Cooperation Foundation Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
WO2020172631A3 (en) * 2019-02-21 2020-10-08 Xencor, Inc. Untargeted and targeted il-10 fc-fusion proteins
WO2020231855A1 (en) 2019-05-10 2020-11-19 Nant Holdings Ip, Llc Nogapendekin alfa-inbakicept for immune stimulant therapies and treatment of viral infections
WO2020234387A1 (en) 2019-05-20 2020-11-26 Cytune Pharma IL-2/IL-15Rßy AGONIST DOSING REGIMENS FOR TREATING CANCER OR INFECTIOUS DISEASES
WO2021006875A1 (en) 2019-07-08 2021-01-14 Nantkwest, Inc. Mononuclear cell derived nk cells
WO2021022304A2 (en) 2019-07-30 2021-02-04 Qlsf Biotherapeutics, Inc. MULTISPECIFIC BINDING COMPOUNDS THAT BIND TO LRRC15 AND CD3ε
WO2021054867A1 (ru) 2019-09-19 2021-03-25 Закрытое Акционерное Общество "Биокад" Иммуноцитокин, включающий гетеро димерный белковый комплекс на основе il-15 и il-15rα
WO2021053559A1 (en) 2019-09-18 2021-03-25 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2021072298A1 (en) 2019-10-11 2021-04-15 Genentech, Inc. Pd-1 targeted il-15/il-15ralpha fc fusion proteins with improved properties
US11059876B2 (en) 2018-02-28 2021-07-13 Pfizer Inc. IL-15 variants and uses thereof
WO2021155042A1 (en) * 2020-01-28 2021-08-05 Genentech, Inc. Il15/il15r alpha heterodimeric fc-fusion proteins for the treatment of cancer
WO2022031876A1 (en) * 2020-08-07 2022-02-10 Genentech, Inc. Flt3 ligand fusion proteins and methods of use
US11267883B2 (en) 2016-12-21 2022-03-08 Cephalon, Inc. Antibodies that specifically bind to human IL-15 and uses thereof
WO2022090203A1 (en) 2020-10-26 2022-05-05 Cytune Pharma IL-2/IL-15Rβү AGONIST FOR TREATING SQUAMOUS CELL CARCINOMA
WO2022090202A1 (en) 2020-10-26 2022-05-05 Cytune Pharma IL-2/IL-15RBβү AGONIST FOR TREATING NON-MELANOMA SKIN CANCER
US11365233B2 (en) 2020-04-10 2022-06-21 Cytomx Therapeutics, Inc. Activatable cytokine constructs and related compositions and methods
US11377477B2 (en) 2018-10-12 2022-07-05 Xencor, Inc. PD-1 targeted IL-15/IL-15RALPHA fc fusion proteins and uses in combination therapies thereof
US11453862B2 (en) 2019-07-08 2022-09-27 Immunitybio, Inc. Mononuclear cell derived NK cells
WO2022232321A1 (en) * 2021-04-28 2022-11-03 Minotaur Therapeutics, Inc. Humanized chimeric bovine antibodies and methods of use
WO2022268991A1 (en) 2021-06-23 2022-12-29 Cytune Pharma Interleukin 15 variants
WO2023017191A1 (en) 2021-08-13 2023-02-16 Cytune Pharma Il-2/il-15rbetagamma agonist combination with antibody-drug conjugates for treating cancer
WO2022268983A3 (en) * 2021-06-23 2023-04-06 Cytune Pharma Interleukin-15 based immunocytokines
WO2023086772A1 (en) 2021-11-12 2023-05-19 Xencor, Inc. Bispecific antibodies that bind to b7h3 and nkg2d
US11667687B2 (en) 2021-03-16 2023-06-06 Cytomx Therapeutics, Inc. Masked activatable interferon constructs
WO2024011179A1 (en) 2022-07-07 2024-01-11 Genentech, Inc. Combinations of il15/il15r alpha heterodimeric fc-fusion proteins and fcrh5xcd3 bispecific antibodies for the treatment of blood cancers
EP4119571A4 (en) * 2020-03-12 2024-04-03 SL Metagen NEW BISPECIFIC PROTEIN AND USE THEREOF
WO2024092038A2 (en) 2022-10-25 2024-05-02 Ablexis, Llc Anti-cd3 antibodies
EP4155316A4 (en) * 2020-05-18 2024-05-08 Shandong Simcere Biopharmaceutical Co., Ltd. HUMAN IL-15 MUTANT AND ITS USE
WO2024102636A1 (en) 2022-11-07 2024-05-16 Xencor, Inc. Bispecific antibodies that bind to b7h3 and mica/b
WO2024107731A2 (en) 2022-11-14 2024-05-23 Ablexis, Llc Anti-pd-l1 antibodies

Families Citing this family (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
WO2014110601A1 (en) 2013-01-14 2014-07-17 Xencor, Inc. Novel heterodimeric proteins
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US10858417B2 (en) * 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
MX2016003292A (es) 2013-09-13 2016-06-24 Beigene Ltd Anticuerpos anti-muerte programada 1 y su uso como terapeuticos y diagnosticos.
US11040111B2 (en) * 2014-03-03 2021-06-22 Academia Sinica Bi-specific antibodies and uses thereof
ME03666B (me) 2014-03-28 2020-10-20 Xencor Inc Bispecifična antitela koja se vezuju za cd38 i cd3
CN106604742B (zh) 2014-07-03 2019-01-11 百济神州有限公司 抗pd-l1抗体及其作为治疗剂及诊断剂的用途
KR20170084326A (ko) 2014-11-26 2017-07-19 젠코어 인코포레이티드 Cd3 및 종양 항원과 결합하는 이종이량체 항체
EP3223907A2 (en) 2014-11-26 2017-10-04 Xencor, Inc. Heterodimeric antibodies that bind cd3 and cd38
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
JP2018512856A (ja) 2015-04-17 2018-05-24 アルパイン イミューン サイエンシズ インコーポレイテッド 調整可能な親和性を有する免疫調節タンパク質
CN108699136B (zh) 2015-12-07 2022-03-18 Xencor股份有限公司 结合cd3和psma的异二聚抗体
EP3443000A2 (en) 2016-04-15 2019-02-20 Alpine Immune Sciences, Inc. Cd80 variant immunomodulatory proteins and uses thereof
MA45037A (fr) 2016-05-18 2019-03-27 Modernatx Inc Polythérapie à base d'arnm pour le traitement du cancer
EP4257613A3 (en) 2016-06-14 2023-12-13 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
EP3475304B1 (en) 2016-06-28 2022-03-23 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
AU2017293423B2 (en) 2016-07-05 2023-05-25 Beigene, Ltd. Combination of a PD-1 antagonist and a RAF inhibitor for treating cancer
US11471488B2 (en) 2016-07-28 2022-10-18 Alpine Immune Sciences, Inc. CD155 variant immunomodulatory proteins and uses thereof
US11834490B2 (en) 2016-07-28 2023-12-05 Alpine Immune Sciences, Inc. CD112 variant immunomodulatory proteins and uses thereof
DK3500299T3 (da) 2016-08-19 2024-01-29 Beigene Switzerland Gmbh Kombination af zanubrutinib med et anti-CD20- eller et anti-PD-1-antistof til anvendelse i behandling af cancer
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
CN110461847B (zh) 2017-01-25 2022-06-07 百济神州有限公司 (S)-7-(1-(丁-2-炔酰基)哌啶-4-基)-2-(4-苯氧基苯基)-4,5,6,7-四氢吡唑并[1,5-a]嘧啶-3-甲酰胺的结晶形式、其制备及用途
AU2018235838B2 (en) * 2017-03-16 2023-12-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
WO2018170023A1 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Pd-l2 variant immunomodulatory proteins and uses thereof
JOP20190260A1 (ar) 2017-05-02 2019-10-31 Merck Sharp & Dohme صيغ ثابتة لأجسام مضادة لمستقبل الموت المبرمج 1 (pd-1) وطرق استخدامها
CA3066518A1 (en) 2017-06-26 2019-01-03 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
MA50360A (fr) 2017-10-10 2020-08-19 Alpine Immune Sciences Inc Protéines immunomodulatrices de variants de ctla-4 et leurs utilisations
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
JP2021502100A (ja) 2017-11-08 2021-01-28 ゼンコア インコーポレイテッド 新規抗pd−1配列を用いた二重特異性および単一特異性抗体
CN111801334B (zh) 2017-11-29 2023-06-09 百济神州瑞士有限责任公司 使用包含btk抑制剂的组合治疗惰性或侵袭性b-细胞淋巴瘤
AU2018390418B2 (en) 2017-12-19 2023-12-21 Xencor, Inc. Engineered IL-2 Fc fusion proteins
EP3773911A2 (en) 2018-04-04 2021-02-17 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
CN110437339B (zh) * 2018-05-04 2021-08-13 免疫靶向有限公司 一种以白介素15为活性成分的融合蛋白型药物前体
EP3794022A1 (en) 2018-05-14 2021-03-24 Werewolf Therapeutics, Inc. Activatable cytokine polypeptides and methods of use thereof
SG11202011349PA (en) 2018-05-14 2020-12-30 Werewolf Therapeutics Inc Activatable interleukin-2 polypeptides and methods of use thereof
JP7477885B2 (ja) 2018-06-22 2024-05-02 キュージーン インコーポレイテッド サイトカインをベースとした生理活性化薬剤およびその使用方法
JP7479383B2 (ja) 2018-09-27 2024-05-08 エクシリオ デベロップメント, インコーポレイテッド マスクされたサイトカインポリペプチド
AU2019355971A1 (en) 2018-10-03 2021-05-06 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
CN109627342B (zh) * 2018-12-10 2022-12-06 苏州近岸蛋白质科技股份有限公司 应用于nk细胞培养的蛋白质、培养基配方组合及制备方法
CA3121813A1 (en) * 2018-12-14 2020-06-18 Proviva Therapeutics (Hong Kong) Limited Il-15 compositions and methods of use thereof
AU2020206269A1 (en) * 2019-01-11 2021-08-05 Memorial Sloan Kettering Cancer Center Multimerization of IL-15/IL-15R-alpha-Fc complexes to enhance immunotherapy
CN111100210B (zh) * 2019-01-30 2022-04-19 武汉九州钰民医药科技有限公司 一种Fc融合蛋白及其应用
WO2020165374A1 (en) 2019-02-14 2020-08-20 Ose Immunotherapeutics Bifunctional molecule comprising il-15ra
CA3132185A1 (en) 2019-03-01 2020-09-10 Xencor, Inc. Heterodimeric antibodies that bind enpp3 and cd3
WO2020200941A1 (en) * 2019-03-29 2020-10-08 F. Hoffmann-La Roche Ag Spr-based binding assay for the functional analysis of multivalent molecules
CA3137512A1 (en) 2019-05-14 2020-11-19 Werewolf Therapeutics, Inc. Separation moieties and methods and use thereof
US11845801B2 (en) 2019-06-12 2023-12-19 AskGene Pharma, Inc. IL-15 prodrugs and methods of use thereof
US20220289838A1 (en) * 2019-08-19 2022-09-15 Nantong Yichen Biopharma. Co. Ltd. Immunocytokine, Preparation for the Same, and Uses Thereof
CN116574183A (zh) * 2019-08-22 2023-08-11 盛禾(中国)生物制药有限公司 多功能抗体、其制备及其用途
CN112409484B (zh) * 2019-08-22 2023-03-21 盛禾(中国)生物制药有限公司 多功能抗体、其制备及其用途
US20210130495A1 (en) * 2019-09-27 2021-05-06 Agenus Inc. Heterodimeric proteins
AU2020358979A1 (en) * 2019-10-03 2022-04-21 Xencor, Inc. Targeted IL-12 heterodimeric Fc-fusion proteins
WO2021092719A1 (zh) * 2019-11-11 2021-05-20 王盛典 一种靶向抗原特异性t细胞诱导其向记忆干性细胞分化的融合蛋白
CN111690068B (zh) * 2019-11-13 2022-04-19 中国科学技术大学 一种IL-15/SuIL-15Rα-dFc-γ4复合体蛋白及其构造方法、应用
CN111690069B (zh) * 2019-11-13 2022-04-19 中国科学技术大学 一种IL-15/SuIL-15Rα-mFc-γ4复合体蛋白及其构造方法、应用
US11692020B2 (en) 2019-11-20 2023-07-04 Anwita Biosciences, Inc. Cytokine fusion proteins, and their pharmaceutical compositions and therapeutic applications
US20230048046A1 (en) * 2019-12-13 2023-02-16 Cugene Inc Novel interleukin-15 (il-15) fusion proteins and uses thereof
CN113321738A (zh) * 2020-02-27 2021-08-31 启愈生物技术(上海)有限公司 肿瘤靶向、抗cd3和t细胞激活三功能融合蛋白及其应用
WO2021188835A1 (en) * 2020-03-18 2021-09-23 City Of Hope Multivalent chemokine receptor binding complexes
WO2021231976A1 (en) 2020-05-14 2021-11-18 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (psma) and cd3
KR102607909B1 (ko) 2020-08-19 2023-12-01 젠코어 인코포레이티드 항-cd28 조성물
EP4208476A1 (en) * 2020-09-01 2023-07-12 Wuxi Biologics Ireland Limited Long-acting il-15 and uses thereof
CN112898436A (zh) * 2020-10-23 2021-06-04 广州医科大学附属肿瘤医院 一种具有靶向功能的鼠pd1和鼠il-15基因融合蛋白的表达及其应用
CN114437228B (zh) * 2020-10-30 2024-02-06 中国科学院生物物理研究所 一种il-2与抗体亚单位构成的双功能融合蛋白
TW202237826A (zh) 2020-11-30 2022-10-01 瑞士商克里斯珀醫療股份公司 基因編輯的自然殺手細胞
WO2022140701A1 (en) 2020-12-24 2022-06-30 Xencor, Inc. Icos targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and icos antigen binding domains
EP4271798A1 (en) 2020-12-30 2023-11-08 CRISPR Therapeutics AG Compositions and methods for differentiating stem cells into nk cells
CN114712494A (zh) * 2021-01-06 2022-07-08 盛禾(中国)生物制药有限公司 一种靶向pd-1的多功能抗体的组合物
CN114712495A (zh) * 2021-01-06 2022-07-08 盛禾(中国)生物制药有限公司 一种多功能抗体的组合物
IL305736A (en) 2021-03-09 2023-11-01 Xencor Inc Heterodimeric antibodies that bind CD3 and CLDN6
EP4305065A1 (en) 2021-03-10 2024-01-17 Xencor, Inc. Heterodimeric antibodies that bind cd3 and gpc3
CN116655771A (zh) * 2021-05-28 2023-08-29 苏州复融生物技术有限公司 一种新型白介素15突变体多肽的开发及其应用
KR20240042442A (ko) 2021-07-28 2024-04-02 제넨테크, 인크. 혈액암 치료를 위한 IL15/IL15R 알파 이종이량체 Fc-융합 단백질
WO2023015198A1 (en) 2021-08-04 2023-02-09 Genentech, Inc. Il15/il15r alpha heterodimeric fc-fusion proteins for the expansion of nk cells in the treatment of solid tumours
KR20230044131A (ko) * 2021-09-24 2023-04-03 바이오엔시스템스 주식회사 Pd-1 및 il-15를 포함하는 융합단백질 이량체 및 이의 용도
KR102595699B1 (ko) 2021-11-03 2023-10-31 주식회사 드론위더스 드론 쉘터
TW202332462A (zh) * 2021-11-18 2023-08-16 大陸商江蘇先聲藥業有限公司 Il-15突變體融合蛋白藥物組合物
US20240025968A1 (en) 2022-04-07 2024-01-25 Xencor, Inc. LAG-3 TARGETED HETERODIMERIC FUSION PROTEINS CONTAINING IL-15/IL-15RA Fc-FUSION PROTEINS AND LAG-3 ANTIGEN BINDING DOMAINS
WO2023222886A1 (en) 2022-05-20 2023-11-23 Depth Charge Ltd Antibody-cytokine fusion proteins
KR20240008802A (ko) * 2022-07-11 2024-01-19 주식회사 지뉴브 사이토카인 융합 단백질
US20240076353A1 (en) * 2022-08-31 2024-03-07 Suzhou Forlong Biotechnology Co., Ltd. Agonistic Interleiukin 15 Complexes and Uses Thereof
CN116178546B (zh) * 2022-10-13 2024-06-18 深圳市百士通科技开发有限公司 一种多功能重组抗体及其制备方法和应用
CN116162171A (zh) * 2022-10-13 2023-05-26 深圳市百士通科技开发有限公司 抗体突变方法在治疗性抗体药物中的应用

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998048032A2 (en) 1997-04-21 1998-10-29 Donlar Corporation POLY-(α-L-ASPARTIC ACID), POLY-(α-L-GLUTAMIC ACID) AND COPOLYMERS OF L-ASP AND L-GLU, METHOD FOR THEIR PRODUCTION AND THEIR USE
US6586207B2 (en) 2000-05-26 2003-07-01 California Institute Of Technology Overexpression of aminoacyl-tRNA synthetases for efficient production of engineered proteins containing amino acid analogues
WO2003073238A2 (en) 2002-02-27 2003-09-04 California Institute Of Technology Computational method for designing enzymes for incorporation of amino acid analogs into proteins
US20040214988A1 (en) 2000-03-23 2004-10-28 California Institute Of Technology Method for stabilization of proteins using non-natural amino acids
WO2005035727A2 (en) 2003-10-09 2005-04-21 Ambrx, Inc. Polymer derivatives
WO2005074524A2 (en) 2004-02-02 2005-08-18 Ambrx, Inc. Modified human interferon polypeptides and their uses
US20120149876A1 (en) 2010-11-05 2012-06-14 Zymeworks Inc. Stable Heterodimeric Antibody Design with Mutations in the Fc Domain
US8216805B2 (en) 1995-03-01 2012-07-10 Genentech, Inc. Knobs and holes heteromeric polypeptides
WO2014145806A2 (en) 2013-03-15 2014-09-18 Xencor, Inc. Heterodimeric proteins
US20140288275A1 (en) 2013-01-14 2014-09-25 Xencor, Inc. Novel heterodimeric proteins
US20150307629A1 (en) 2014-03-28 2015-10-29 Matthew Bernett Bispecific antibodies that bind to CD38 and CD3
AU2014377106A1 (en) * 2014-01-08 2016-08-11 Jiangsu Hengrui Medicine Co., Ltd. IL-15 heterodimeric protein and uses thereof
US20160244525A1 (en) 2014-11-05 2016-08-25 Genentech, Inc. Anti-fgfr2/3 antibodies and methods using same
WO2016141387A1 (en) * 2015-03-05 2016-09-09 Xencor, Inc. Modulation of t cells with bispecific antibodies and fc fusions

Family Cites Families (371)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
CU22545A1 (es) 1994-11-18 1999-03-31 Centro Inmunologia Molecular Obtención de un anticuerpo quimérico y humanizado contra el receptor del factor de crecimiento epidérmico para uso diagnóstico y terapéutico
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4169888A (en) 1977-10-17 1979-10-02 The Upjohn Company Composition of matter and process
US4307016A (en) 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
US4256746A (en) 1978-11-14 1981-03-17 Takeda Chemical Industries Dechloromaytansinoids, their pharmaceutical compositions and method of use
JPS55102583A (en) 1979-01-31 1980-08-05 Takeda Chem Ind Ltd 20-acyloxy-20-demethylmaytansinoid compound
JPS55162791A (en) 1979-06-05 1980-12-18 Takeda Chem Ind Ltd Antibiotic c-15003pnd and its preparation
JPS6023084B2 (ja) 1979-07-11 1985-06-05 味の素株式会社 代用血液
JPS5645483A (en) 1979-09-19 1981-04-25 Takeda Chem Ind Ltd C-15003phm and its preparation
EP0028683A1 (en) 1979-09-21 1981-05-20 Takeda Chemical Industries, Ltd. Antibiotic C-15003 PHO and production thereof
JPS5645485A (en) 1979-09-21 1981-04-25 Takeda Chem Ind Ltd Production of c-15003pnd
US4364935A (en) 1979-12-04 1982-12-21 Ortho Pharmaceutical Corporation Monoclonal antibody to a human prothymocyte antigen and methods of preparing same
WO1982001188A1 (en) 1980-10-08 1982-04-15 Takeda Chemical Industries Ltd 4,5-deoxymaytansinoide compounds and process for preparing same
US4450254A (en) 1980-11-03 1984-05-22 Standard Oil Company Impact improvement of high nitrile resins
US4313946A (en) 1981-01-27 1982-02-02 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids from Trewia nudiflora
US4315929A (en) 1981-01-27 1982-02-16 The United States Of America As Represented By The Secretary Of Agriculture Method of controlling the European corn borer with trewiasine
JPS57192389A (en) 1981-05-20 1982-11-26 Takeda Chem Ind Ltd Novel maytansinoid
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4943533A (en) 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
US4970198A (en) 1985-10-17 1990-11-13 American Cyanamid Company Antitumor antibiotics (LL-E33288 complex)
EP0206448B1 (en) 1985-06-19 1990-11-14 Ajinomoto Co., Inc. Hemoglobin combined with a poly(alkylene oxide)
JPS63502716A (ja) 1986-03-07 1988-10-13 マサチューセッツ・インステチュート・オブ・テクノロジー 糖タンパク安定性の強化方法
DE3783588T2 (de) 1986-04-17 1993-06-09 Kyowa Hakko Kogyo Kk Neue verbindungen dc-88a und dc-89a1 und deren herstellungsverfahren.
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US4880935A (en) 1986-07-11 1989-11-14 Icrf (Patents) Limited Heterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
US5053394A (en) 1988-09-21 1991-10-01 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
JP3040121B2 (ja) 1988-01-12 2000-05-08 ジェネンテク,インコーポレイテッド 増殖因子レセプターの機能を阻害することにより腫瘍細胞を処置する方法
IL106992A (en) 1988-02-11 1994-06-24 Bristol Myers Squibb Co Noble hydrazonic history of anthracycline and methods for their preparation
US5084468A (en) 1988-08-11 1992-01-28 Kyowa Hakko Kogyo Co., Ltd. Dc-88a derivatives
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
JP2598116B2 (ja) 1988-12-28 1997-04-09 協和醗酵工業株式会社 新規物質dc113
JP2510335B2 (ja) 1989-07-03 1996-06-26 協和醗酵工業株式会社 Dc―88a誘導体
US5187186A (en) 1989-07-03 1993-02-16 Kyowa Hakko Kogyo Co., Ltd. Pyrroloindole derivatives
CA2026147C (en) 1989-10-25 2006-02-07 Ravi J. Chari Cytotoxic agents comprising maytansinoids and their therapeutic use
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
AU652936B2 (en) 1990-05-07 1994-09-15 Scripps Clinic And Research Foundation Intermediates in the formation of the calicheamicin and esperamicin oligosaccharides
US5968509A (en) 1990-10-05 1999-10-19 Btp International Limited Antibodies with binding affinity for the CD3 antigen
CZ282603B6 (cs) 1991-03-06 1997-08-13 Merck Patent Gesellschaft Mit Beschränkter Haftun G Humanizované a chimerické monoklonální protilátky
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
CA2103059C (en) 1991-06-14 2005-03-22 Paul J. Carter Method for making humanized antibodies
US5264586A (en) 1991-07-17 1993-11-23 The Scripps Research Institute Analogs of calicheamicin gamma1I, method of making and using the same
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
GB9206422D0 (en) 1992-03-24 1992-05-06 Bolt Sarah L Antibody preparation
EP0563475B1 (en) 1992-03-25 2000-05-31 Immunogen Inc Cell binding agent conjugates of derivatives of CC-1065
ZA932522B (en) 1992-04-10 1993-12-20 Res Dev Foundation Immunotoxins directed against c-erbB-2(HER/neu) related surface antigens
US6329507B1 (en) 1992-08-21 2001-12-11 The Dow Chemical Company Dimer and multimer forms of single chain polypeptides
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
ATE199392T1 (de) 1992-12-04 2001-03-15 Medical Res Council Multivalente und multispezifische bindungsproteine, deren herstellung und verwendung
SG55079A1 (en) 1992-12-11 1998-12-21 Dow Chemical Co Multivalent single chain antibodies
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
EP0731106B1 (en) 1993-10-01 2004-11-17 Teikoku Hormone Mfg. Co., Ltd. Dolastatin derivatives
GB9401182D0 (en) 1994-01-21 1994-03-16 Inst Of Cancer The Research Antibodies to EGF receptor and their antitumour effect
ES2220927T3 (es) 1994-04-22 2004-12-16 Kyowa Hakko Kogyo Co., Ltd. Derivados de dc-89.
JPH07309761A (ja) 1994-05-20 1995-11-28 Kyowa Hakko Kogyo Co Ltd デュオカルマイシン誘導体の安定化法
US5945311A (en) 1994-06-03 1999-08-31 GSF--Forschungszentrumfur Umweltund Gesundheit Method for producing heterologous bi-specific antibodies
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5550246A (en) 1994-09-07 1996-08-27 The Scripps Research Institute Calicheamicin mimics
US5541087A (en) 1994-09-14 1996-07-30 Fuji Immunopharmaceuticals Corporation Expression and export technology of proteins as immunofusins
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
EP0831880A4 (en) 1995-06-07 2004-12-01 Imclone Systems Inc ANTIBODIES AND FRAGMENTS OF ANTIBODIES INHIBITING TUMOR GROWTH
US7696338B2 (en) 1995-10-30 2010-04-13 The United States Of America As Represented By The Department Of Health And Human Services Immunotoxin fusion proteins and means for expression thereof
WO1997023243A1 (en) 1995-12-22 1997-07-03 Bristol-Myers Squibb Company Branched hydrazone linkers
US6451308B1 (en) 1996-04-26 2002-09-17 Beth Israel Deaconess Medical Center Antagonists of interleukin-15
WO1997041232A1 (en) 1996-04-26 1997-11-06 Beth Israel Deaconess Medical Center Antagonists of interleukin-15
DE69830901T2 (de) 1997-05-02 2006-05-24 Genentech Inc., San Francisco ein verfahren zur herstellung multispezifischer antikörper die heteromultimere und gemeinsame komponenten besitzen
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
DE69922159T2 (de) 1998-01-23 2005-12-01 Vlaams Interuniversitair Instituut Voor Biotechnologie Mehrzweck-antikörperderivate
ATE458007T1 (de) 1998-04-20 2010-03-15 Glycart Biotechnology Ag Glykosylierungs-engineering von antikörpern zur verbesserung der antikörperabhängigen zellvermittelten zytotoxizität
IL138857A0 (en) 1998-04-21 2001-10-31 Micromet Ges For Biomedizinisc Cd19xcd3 specific polypeptides and uses thereof
US6455677B1 (en) 1998-04-30 2002-09-24 Boehringer Ingelheim International Gmbh FAPα-specific antibody with improved producibility
AU760854B2 (en) 1998-06-22 2003-05-22 Immunomedics Inc. Use of bi-specific antibodies for pre-targeting diagnosis and therapy
GB9815909D0 (en) 1998-07-21 1998-09-16 Btg Int Ltd Antibody preparation
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US6723538B2 (en) 1999-03-11 2004-04-20 Micromet Ag Bispecific antibody and chemokine receptor constructs
CA2704600C (en) 1999-04-09 2016-10-25 Kyowa Hakko Kirin Co., Ltd. A method for producing antibodies with increased adcc activity
US6939545B2 (en) 1999-04-28 2005-09-06 Genetics Institute, Llc Composition and method for treating inflammatory disorders
MXPA02001417A (es) 1999-08-09 2002-08-12 Lexigen Pharm Corp Complejos multiples de citosina-anticuerpo.
AU775373B2 (en) 1999-10-01 2004-07-29 Immunogen, Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
US7303749B1 (en) 1999-10-01 2007-12-04 Immunogen Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
JP4668498B2 (ja) 1999-10-19 2011-04-13 協和発酵キリン株式会社 ポリペプチドの製造方法
US6716410B1 (en) 1999-10-26 2004-04-06 The Regents Of The University Of California Reagents and methods for diagnosing, imaging and treating atherosclerotic disease
JP2003523771A (ja) 2000-02-25 2003-08-12 ザ ガバメント オブ ザ ユナイテッド ステイツ, アズ レプレゼンテッド バイ ザ セクレタリー オブ ザ デパートメント オブ ヘルス アンド ヒューマン サービシーズ 改善された細胞傷害性および収率を有する抗EGFRvIIIscFv、それに基づく免疫毒素、ならびにその使用方法
US20010035606A1 (en) 2000-03-28 2001-11-01 Schoen Alan H. Set of blocks for packing a cube
WO2001083525A2 (en) 2000-05-03 2001-11-08 Amgen Inc. Modified peptides, comprising an fc domain, as therapeutic agents
KR100480985B1 (ko) 2000-05-19 2005-04-07 이수화학 주식회사 표피 성장 인자 수용체에 대한 사람화된 항체
EP1299419A2 (en) 2000-05-24 2003-04-09 Imclone Systems, Inc. Bispecific immunoglobulin-like antigen binding proteins and method of production
JP2004523205A (ja) 2000-07-25 2004-08-05 イムノメディクス, インコーポレイテッド 多価標的結合タンパク質
US6333410B1 (en) 2000-08-18 2001-12-25 Immunogen, Inc. Process for the preparation and purification of thiol-containing maytansinoids
DE10043437A1 (de) 2000-09-04 2002-03-28 Horst Lindhofer Verwendung von trifunktionellen bispezifischen und trispezifischen Antikörpern zur Behandlung von malignem Aszites
CA2424977C (en) 2000-10-06 2008-03-18 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
HU231090B1 (hu) 2000-10-06 2020-07-28 Kyowa Kirin Co., Ltd. Antitest-kompozíciót termelő sejt
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20020147312A1 (en) 2001-02-02 2002-10-10 O'keefe Theresa Hybrid antibodies and uses thereof
EP1243276A1 (en) 2001-03-23 2002-09-25 Franciscus Marinus Hendrikus De Groot Elongated and multiple spacers containing activatible prodrugs
EP1383800A4 (en) 2001-04-02 2004-09-22 Idec Pharma Corp RECOMBINANT ANTIBODIES CO-EXPRESSED WITH GNTIII
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
MXPA03011094A (es) 2001-05-31 2004-12-06 Medarex Inc Citotoxinas, profarmacos, ligadores, y estabilizadores utiles para ello.
CN100497389C (zh) 2001-06-13 2009-06-10 根马布股份公司 表皮生长因子受体(egfr)的人单克隆抗体
EP1411983A4 (en) 2001-06-26 2006-06-21 Imclone Systems Inc BISPECIFIC ANTIBODIES BINDING TO VEGF RECEPTORS
HUP0600342A3 (en) 2001-10-25 2011-03-28 Genentech Inc Glycoprotein compositions
US20080219974A1 (en) 2002-03-01 2008-09-11 Bernett Matthew J Optimized antibodies that target hm1.24
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US7332580B2 (en) 2002-04-05 2008-02-19 The Regents Of The University Of California Bispecific single chain Fv antibody molecules and methods of use thereof
KR101086533B1 (ko) 2002-05-24 2011-11-23 쉐링 코포레이션 중화 사람 항-igfr 항체, 이를 제조하는 방법 및 이를 포함하는 조성물
AU2003263964C1 (en) 2002-07-31 2010-08-19 Seagen Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US20060235208A1 (en) 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
US7820166B2 (en) 2002-10-11 2010-10-26 Micromet Ag Potent T cell modulating molecules
AU2003282624A1 (en) 2002-11-14 2004-06-03 Syntarga B.V. Prodrugs built as multiple self-elimination-release spacers
EP1578397B1 (en) 2002-11-15 2012-12-26 Genmab A/S Human monoclonal antibodies against cd25
WO2004063351A2 (en) 2003-01-09 2004-07-29 Macrogenics, Inc. IDENTIFICATION AND ENGINEERING OF ANTIBODIES WITH VARIANT Fc REGIONS AND METHODS OF USING SAME
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US7610156B2 (en) 2003-03-31 2009-10-27 Xencor, Inc. Methods for rational pegylation of proteins
PT3524611T (pt) 2003-05-20 2021-04-01 Immunogen Inc Agentes citotóxicos melhorados compreendendo novos maitansinóides
US7276497B2 (en) 2003-05-20 2007-10-02 Immunogen Inc. Cytotoxic agents comprising new maytansinoids
WO2004106381A1 (en) 2003-05-31 2004-12-09 Micromet Ag Pharmaceutical compositions comprising bispecific anti-cd3, anti-cd19 antibody constructs for the treatment of b-cell related disorders
RU2005137325A (ru) 2003-05-31 2006-09-10 Микромет Аг (De) Фармацевтическая композиция, содержащая конструкт, специфичный к ерсам
US7888134B2 (en) 2003-06-05 2011-02-15 Oakland University Immunosensors: scFv-linker design for surface immobilization
WO2005014642A2 (en) 2003-07-21 2005-02-17 Transgene S.A. Novel multifunctional cytokines
US20060134105A1 (en) 2004-10-21 2006-06-22 Xencor, Inc. IgG immunoglobulin variants with optimized effector function
US20150071948A1 (en) 2003-09-26 2015-03-12 Gregory Alan Lazar Novel immunoglobulin variants
US20050176028A1 (en) 2003-10-16 2005-08-11 Robert Hofmeister Deimmunized binding molecules to CD3
SI1725249T1 (sl) 2003-11-06 2014-04-30 Seattle Genetics, Inc. Spojine monometilvalina, sposobne konjugacije na ligande
US20050142133A1 (en) 2003-12-03 2005-06-30 Xencor, Inc. Optimized proteins that target the epidermal growth factor receptor
WO2005056759A2 (en) 2003-12-04 2005-06-23 Xencor, Inc. Methods of generating variant proteins with increased host string content and compositions thereof
PL1718677T3 (pl) 2003-12-19 2012-09-28 Genentech Inc Jednowartościowe fragmenty przeciwciała stosowane jako środki lecznicze
US7235641B2 (en) 2003-12-22 2007-06-26 Micromet Ag Bispecific antibodies
EP1718670B1 (en) 2004-02-27 2011-07-13 INSERM (Institut National de la Santé et de la Recherche Médicale) Il-15 binding site for il 15-ralpha and specific il-15 mutants having agonist/antagonist activity
WO2005092925A2 (en) 2004-03-24 2005-10-06 Xencor, Inc. Immunoglobulin variants outside the fc region
AU2005244980B2 (en) 2004-05-19 2011-09-15 E. R. Squibb & Sons, L.L.C. Chemical linkers and conjugates thereof
RU2402548C2 (ru) 2004-05-19 2010-10-27 Медарекс, Инк. Химические линкеры и их конъюгаты
BRPI0511782B8 (pt) 2004-06-03 2021-05-25 Novimmune Sa anticorpos anti-cd3, uso e método de produção dos mesmos, composição farmacêutica, molécula de ácido nucleico isolada e vetor
US20060018897A1 (en) 2004-06-28 2006-01-26 Transtarget Inc. Bispecific antibodies
JP2008512352A (ja) 2004-07-17 2008-04-24 イムクローン システムズ インコーポレイティド 新規な四価の二重特異性抗体
AU2005282720B2 (en) 2004-09-02 2011-08-04 Genentech, Inc. Anti-FC-gamma RIIB receptor antibody and uses therefor
CU23472A1 (es) 2004-09-17 2009-12-17 Ct Ingenieria Genetica Biotech Péptido antagonista de la interleucina-15
WO2006034488A2 (en) 2004-09-23 2006-03-30 Genentech, Inc. Cysteine engineered antibodies and conjugates
CA2580796C (en) 2004-09-24 2013-03-26 Amgen Inc. Modified fc molecules having peptides inserted in internal loop regions
AU2005335714B2 (en) 2004-11-10 2012-07-26 Macrogenics, Inc. Engineering Fc antibody regions to confer effector function
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US8066989B2 (en) 2004-11-30 2011-11-29 Trion Pharma Gmbh Method of treating tumor growth and metastasis by using trifunctional antibodies to reduce the risk for GvHD in allogeneic antitumor cell therapy
AU2005315658A1 (en) 2004-12-13 2006-06-22 Cytos Biotechnology Ag IL-15 antigen arrays and uses thereof
DE602006003695D1 (de) 2005-01-05 2009-01-02 F Star Biotech Forsch & Entw Synthetische Immunoglobulindomänen mit in Regionen des Moleküls, die sich von den die Komplementarität bestimmenden Bereichen unterscheiden, modifizierten Bindeeigenschaften
JP4986633B2 (ja) 2005-01-12 2012-07-25 協和発酵キリン株式会社 安定化されたヒトIgG2およびIgG3抗体
EP3623473A1 (en) 2005-03-31 2020-03-18 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
US7714016B2 (en) 2005-04-08 2010-05-11 Medarex, Inc. Cytotoxic compounds and conjugates with cleavable substrates
US20060257361A1 (en) 2005-04-12 2006-11-16 Government Of The Us, As Represented By The Secretary, Department Of Health And Human Services Novel form of interleukin-15, Fc-IL-15, and methods of use
US9284375B2 (en) 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
MX2007014474A (es) 2005-05-17 2008-02-07 Univ Connecticut Composiciones y metodos para inmunomodulacion en un organismo.
US8309690B2 (en) 2005-07-01 2012-11-13 Medimmune, Llc Integrated approach for generating multidomain protein therapeutics
US9017992B2 (en) 2005-07-01 2015-04-28 Dako Denmark A/S In situ hybridization detection method
PT2298815E (pt) 2005-07-25 2015-07-16 Emergent Product Dev Seattle Moléculas de ligaçao especificas para cd37 e especificas para cd20
RU2421242C2 (ru) 2005-07-25 2011-06-20 Трабьон Фармасьютикалз, Инк. Применение однократной дозы cd20-специфических связывающих молекул
CA2617907A1 (en) 2005-08-05 2007-02-15 Syntarga B.V. Triazole-containing releasable linkers and conjugates comprising the same
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
UA92505C2 (ru) 2005-09-12 2010-11-10 Новиммюн С.А. Композиции на основе антитела против cd3
DK1931709T3 (en) 2005-10-03 2017-03-13 Xencor Inc FC VARIETIES WITH OPTIMIZED FC RECEPTOR BINDING PROPERTIES
AU2006301492B2 (en) 2005-10-11 2011-06-09 Amgen Research (Munich) Gmbh Compositions comprising cross-species-specific antibodies and uses thereof
JP2009511067A (ja) 2005-10-14 2009-03-19 メディミューン,エルエルシー 抗体ライブラリーの細胞提示
WO2007047829A2 (en) 2005-10-19 2007-04-26 Laboratoires Serono S.A. Novel heterodimeric proteins and uses thereof
EP1777294A1 (en) 2005-10-20 2007-04-25 Institut National De La Sante Et De La Recherche Medicale (Inserm) IL-15Ralpha sushi domain as a selective and potent enhancer of IL-15 action through IL-15Rbeta/gamma, and hyperagonist (IL15Ralpha sushi -IL15) fusion proteins
TW200732350A (en) 2005-10-21 2007-09-01 Amgen Inc Methods for generating monovalent IgG
WO2007059404A2 (en) 2005-11-10 2007-05-24 Medarex, Inc. Duocarmycin derivatives as novel cytotoxic compounds and conjugates
WO2007062037A2 (en) 2005-11-21 2007-05-31 Laboratoires Serono Sa Compositions and methods of producing hybrid antigen binding molecules and uses thereof
CA2633713A1 (en) 2005-12-21 2007-06-28 Micromet Ag Epha2 bite molecules and uses thereof
NZ596494A (en) 2006-01-13 2013-07-26 Us Gov Nat Inst Health Codon optimized il-15 and il-15r-alpha genes for expression in mammalian cells
US8940784B2 (en) 2006-02-02 2015-01-27 Syntarga B.V. Water-soluble CC-1065 analogs and their conjugates
EP1820513A1 (en) 2006-02-15 2007-08-22 Trion Pharma Gmbh Destruction of tumor cells expressing low to medium levels of tumor associated target antigens by trifunctional bispecific antibodies
EP1829895A1 (en) 2006-03-03 2007-09-05 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Bispecific molecule binding TLR9 and CD32 and comprising a T cell epitope for treatment of allergies
NZ591252A (en) 2006-03-17 2012-06-29 Biogen Idec Inc Methods of designing antibody or antigen binding fragments thereof with substituted non-covarying amino acids
PT1999154E (pt) 2006-03-24 2013-01-24 Merck Patent Gmbh Domínios proteicos heterodiméricos modificados
JP5624276B2 (ja) 2006-03-31 2014-11-12 中外製薬株式会社 抗体の血中動態を制御する方法
US9670269B2 (en) 2006-03-31 2017-06-06 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
EP2007423A2 (en) 2006-04-05 2008-12-31 Pfizer Products Incorporated Ctla4 antibody combination therapy
EP2016098B1 (en) 2006-05-08 2016-09-07 Philogen S.p.A. Antibody-targeted cytokines for therapy
WO2007147901A1 (en) 2006-06-22 2007-12-27 Novo Nordisk A/S Production of bispecific antibodies
AT503889B1 (de) 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F Multivalente immunglobuline
AT503902B1 (de) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw Verfahren zur manipulation von immunglobulinen
AT503861B1 (de) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw Verfahren zur manipulation von t-zell-rezeptoren
JP5473603B2 (ja) 2006-10-02 2014-04-16 シー レーン バイオテクノロジーズ, エルエルシー 多様な合成ペプチドおよびポリペプチドライブラリーの設計および構築
MX2009009912A (es) 2007-03-27 2010-01-18 Sea Lane Biotechnologies Llc Constructos y colecciones que comprenden secuencias de cadena ligera sustitutas de anticuerpos.
EP1975178A1 (en) 2007-03-30 2008-10-01 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Transcytotic modular antibody
MX2009010611A (es) 2007-04-03 2010-03-26 Micromet Ag Enlazadores biespecificos, especificos para especies.
PT2520590T (pt) 2007-04-03 2018-11-14 Amgen Res Munich Gmbh Domínio de ligação específico de espécies cruzadas
WO2008124858A2 (en) 2007-04-11 2008-10-23 F-Star Biotechnologische Forschungs- Und Entwicklungsges. M.B.H. Targeted receptor
EP2155789B1 (en) 2007-05-01 2013-07-24 Research Development Foundation Immunoglobulin fc libraries
PT2160401E (pt) 2007-05-11 2014-10-30 Altor Bioscience Corp Moléculas de fusão e variantes de il-15
BRPI0811857A2 (pt) 2007-05-14 2014-10-21 Biogen Idec Inc Regiões fc (scfc) de cadeia simples, polipeptídeos de aglutinação que as compreendem e métodos relacionados.
ES2659517T3 (es) 2007-05-30 2018-03-16 Xencor, Inc. Métodos y composiciones para inhibir células que expresan CD32B
CA2688275A1 (en) 2007-05-31 2008-12-04 Genmab A/S Stable igg4 antibodies
BRPI0812562A2 (pt) 2007-06-12 2019-09-24 Trubion Pharmaceuticals Inc composições terapêuticas anti-cd20 e métodos
ES2627223T3 (es) 2007-06-26 2017-07-27 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H Presentación de agentes de unión
ES2466916T3 (es) 2007-06-27 2014-06-11 Admune Therapeutics Llc Complejos de IL-15 e IL-15R alfa y usos de los mismos
AU2008282729B2 (en) 2007-08-01 2015-07-16 Scott & White Healthcare A fold-back diabody diphtheria toxin immunotoxin and methods of use
US8680293B2 (en) 2007-08-01 2014-03-25 Syntarga B.V. Substituted CC-1065 analogs and their conjugates
AU2008296386A1 (en) 2007-08-28 2009-03-12 Biogen Idec Ma Inc. Compositions that bind multiple epitopes of IGF-1R
EP2033657A1 (de) 2007-09-04 2009-03-11 Trion Pharma Gmbh Intraoperative trifunktionale Antikörper-Applikation zur Prophylaxe intraperitonealer Tumorzelldissemination
EP2197911A2 (en) 2007-09-14 2010-06-23 Amgen Inc. Homogeneous antibody populations
ES2622460T3 (es) 2007-09-26 2017-07-06 Ucb Biopharma Sprl Fusiones de anticuerpos con doble especificidad
ES2566957T3 (es) 2007-09-26 2016-04-18 Chugai Seiyaku Kabushiki Kaisha Región constante de anticuerpo modificado
EP3689912A1 (en) 2007-09-26 2020-08-05 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
EP3575317A1 (en) 2007-12-26 2019-12-04 Xencor, Inc. Fc variants with altered binding to fcrn
DK2235064T3 (en) 2008-01-07 2016-01-11 Amgen Inc A process for the preparation of heterodimeric Fc molecules using electrostatic control effects
WO2009106096A1 (en) 2008-02-27 2009-09-03 Fresenius Biotech Gmbh Treatment of resistant tumors with trifunctional antibodies
WO2009126944A1 (en) 2008-04-11 2009-10-15 Trubion Pharmaceuticals, Inc. Cd37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
US8314213B2 (en) 2008-04-18 2012-11-20 Xencor, Inc. Human equivalent monoclonal antibodies engineered from nonhuman variable regions
CN102076865B (zh) 2008-05-02 2016-03-16 西雅图基因公司 用于制造核心岩藻糖基化降低的抗体和抗体衍生物的方法和组合物
PE20100092A1 (es) 2008-06-03 2010-03-12 Abbott Lab Inmunoglobulina con dominio variable dual y usos de la misma
WO2010017103A2 (en) 2008-08-04 2010-02-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Servic Fully human anti-human nkg2d monoclonal antibodies
WO2010028796A1 (en) 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Trispecific hexavalent antibodies
EP2703415B1 (en) 2008-09-17 2019-03-06 Xencor, Inc. Compositions and methods for treating IgE-mediated disorders
US20170247470A9 (en) 2008-09-17 2017-08-31 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
KR20110047255A (ko) 2008-09-26 2011-05-06 로슈 글리카트 아게 이중특이적 항-egfr/항-igf-1r 항체
EP2352763B2 (en) 2008-10-01 2022-09-21 Amgen Research (Munich) GmbH Bispecific single chain antibodies with specificity for high molecular weight target antigens
EP2352765B1 (en) 2008-10-01 2018-01-03 Amgen Research (Munich) GmbH Cross-species-specific single domain bispecific single chain antibody
CA2738565C (en) 2008-10-01 2023-10-10 Micromet Ag Cross-species-specific psmaxcd3 bispecific single chain antibody
EP2370467B1 (en) 2008-10-01 2016-09-07 Amgen Research (Munich) GmbH Cross-species-specific pscaxcd3, cd19xcd3, c-metxcd3, endosialinxcd3, epcamxc d3, igf-1rxcd3 or fapalpha xcd3 bispecific single chain antibody
SG172754A1 (en) 2008-10-10 2011-08-29 Trubion Pharmaceuticals Inc Tcr complex immunotherapeutics
HUE035798T2 (en) 2008-11-03 2018-05-28 Syntarga Bv CC-1065 analogues and conjugates
US8057507B2 (en) 2009-01-16 2011-11-15 Novate Medical Limited Vascular filter
JP2012515556A (ja) 2009-01-23 2012-07-12 バイオジェン・アイデック・エムエイ・インコーポレイテッド 低下したエフェクタ機能を有する安定化Fcポリペプチドおよび使用方法
MX2011009306A (es) 2009-03-06 2011-10-13 Genentech Inc Formulacion con anticuerpo.
EP2233500A1 (en) 2009-03-20 2010-09-29 LFB Biotechnologies Optimized Fc variants
MX2011010159A (es) 2009-04-02 2011-10-17 Roche Glycart Ag Anticuerpos multiespecificos que comprenden anticuerpos de longitud completa y fragmentos fab de cadena sencilla.
JP5616428B2 (ja) 2009-04-07 2014-10-29 ロシュ グリクアート アクチェンゲゼルシャフト 三価の二重特異性抗体
CN102361883A (zh) 2009-04-07 2012-02-22 罗氏格黎卡特股份公司 双特异性抗-ErbB-1/抗-c-Met抗体
EP2417159A1 (en) 2009-04-07 2012-02-15 Roche Glycart AG Bispecific anti-erbb-3/anti-c-met antibodies
EP2241576A1 (en) 2009-04-17 2010-10-20 Trion Pharma Gmbh Use of trifunctional bispecific antibodies for the treatment of tumors associated with CD133+/EpCAM+ cancer stem cells
EP2435473B1 (en) 2009-05-27 2013-10-02 F.Hoffmann-La Roche Ag Tri- or tetraspecific antibodies
CA2764729C (en) 2009-06-26 2019-04-02 Sea Lane Biotechnologies, Llc Expression of surrogate light chains
EP2445936A1 (en) 2009-06-26 2012-05-02 Regeneron Pharmaceuticals, Inc. Readily isolated bispecific antibodies with native immunoglobulin format
CA2766065C (en) 2009-06-30 2020-07-21 Research Development Foundation Immunoglobulin fc polypeptides
US9308258B2 (en) 2009-07-08 2016-04-12 Amgen Inc. Stable and aggregation free antibody FC molecules through CH3 domain interface engineering
JP2013501817A (ja) 2009-08-14 2013-01-17 アメリカ合衆国 胸腺産出の増大およびリンパ球減少症の治療のためのil−15の使用
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
DE102009045006A1 (de) 2009-09-25 2011-04-14 Technische Universität Dresden Anti-CD33 Antikörper und ihre Anwendung zum Immunotargeting bei der Behandlung von CD33-assoziierten Erkrankungen
ES2688978T3 (es) 2009-11-23 2018-11-07 Amgen Inc. Anticuerpo monomérico Fc
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
CN102711810B (zh) 2009-11-30 2015-04-22 詹森生物科技公司 效应子功能已消除的抗体Fc区突变体
MX2012007497A (es) 2009-12-25 2012-08-01 Chugai Pharmaceutical Co Ltd Metodo para modificacion de polipeptidos para purificar multimeros de polipeptido.
ME02505B (me) 2009-12-29 2017-02-20 Aptevo Res & Development Llc Heterodimerni vezujući proteini i njihove upotrebe
US20130129723A1 (en) 2009-12-29 2013-05-23 Emergent Product Development Seattle, Llc Heterodimer Binding Proteins and Uses Thereof
US20110189178A1 (en) 2010-02-04 2011-08-04 Xencor, Inc. Immunoprotection of Therapeutic Moieties Using Enhanced Fc Regions
WO2011120134A1 (en) 2010-03-29 2011-10-06 Zymeworks, Inc. Antibodies with enhanced or suppressed effector function
TWI667257B (zh) 2010-03-30 2019-08-01 中外製藥股份有限公司 促進抗原消失之具有經修飾的FcRn親和力之抗體
US9150663B2 (en) 2010-04-20 2015-10-06 Genmab A/S Heterodimeric antibody Fc-containing proteins and methods for production thereof
WO2011133886A2 (en) 2010-04-23 2011-10-27 Genentech, Inc. Production of heteromultimeric proteins
EP2569337A1 (en) 2010-05-14 2013-03-20 Rinat Neuroscience Corp. Heterodimeric proteins and methods for producing and purifying them
WO2011154453A1 (en) 2010-06-09 2011-12-15 Genmab A/S Antibodies against human cd38
CA2802344C (en) 2010-06-18 2023-06-13 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions
CN103052649B (zh) 2010-07-29 2015-12-16 Xencor公司 具有修改的等电点的抗体
MX339622B (es) 2010-08-02 2016-06-02 Macrogenics Inc Diacuerpos covalentes y sus usos.
CN103068846B9 (zh) 2010-08-24 2016-09-28 弗·哈夫曼-拉罗切有限公司 包含二硫键稳定性Fv片段的双特异性抗体
WO2012032080A1 (en) 2010-09-07 2012-03-15 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H Stabilised human fc
JP6251570B2 (ja) 2010-09-21 2017-12-20 アルター・バイオサイエンス・コーポレーション 多量体il−15可溶性融合分子並びにその製造及び使用方法
CN103533943B (zh) 2010-11-10 2018-02-13 安进研发(慕尼黑)股份有限公司 由cd3特异性结合结构域导致的不良作用的预防
EP3075745B1 (en) 2011-02-10 2018-09-05 Roche Glycart AG Mutant interleukin-2 polypeptides
ES2676878T3 (es) 2011-03-03 2018-07-25 Zymeworks Inc. Diseño de armazón de heteromultímero multivalente y constructos
MX2013010172A (es) 2011-03-11 2013-10-25 Amgen Inc Metodo de analisis mutacional correlacionado para mejorar anticuerpos terapeuticos.
CA2830254C (en) 2011-03-16 2019-09-10 Amgen Inc. Fc variants
BR112013023918A2 (pt) 2011-03-25 2016-12-13 Glenmark Pharmaceuticals Sa imunoglobulina hetero-dimérica ou fragmento hetero-dimérico da mesma, método para produzir uma imunoglobulina hetero-dimérica ou fragmento hetero-dimérico da mesma, método para construir uma interface proteína-proteína de um domínio de uma proteína de múltiplos domínios e uso de um domínio doador de um primeiro e de um segundo membro de uma super-família de imunoglobulina de ocorrência natural
TWI588156B (zh) 2011-03-28 2017-06-21 賽諾菲公司 具有交叉結合區定向之雙重可變區類抗體結合蛋白
KR20160044598A (ko) 2011-03-29 2016-04-25 로슈 글리카트 아게 항체 Fc 변이체
AU2012247762B2 (en) 2011-04-28 2017-07-06 Amgen Research (Munich) Gmbh Dosage regimen for administering a CD19xCD3 bispecific antibody to patients at risk for potential adverse effects
EA201892619A1 (ru) 2011-04-29 2019-04-30 Роше Гликарт Аг Иммуноконъюгаты, содержащие мутантные полипептиды интерлейкина-2
ME03440B (me) 2011-05-21 2020-01-20 Macrogenics Inc Cd3-vezujući molekuli sposobni za vezivanje za humani i nehumani cd3
AU2012258907A1 (en) 2011-05-25 2013-11-07 Merck Sharp & Dohme Corp. Method for preparing Fc-containing polypeptides having improved properties
EP2537933A1 (en) 2011-06-24 2012-12-26 Institut National de la Santé et de la Recherche Médicale (INSERM) An IL-15 and IL-15Ralpha sushi domain based immunocytokines
DK2728002T3 (da) 2011-06-30 2022-04-04 Chugai Pharmaceutical Co Ltd Heterodimeriseret polypeptid
US20140155581A1 (en) 2011-07-06 2014-06-05 Medimmune, Llc Methods For Making Multimeric Polypeptides
EP2736928B1 (en) 2011-07-28 2019-01-09 i2 Pharmaceuticals, Inc. Sur-binding proteins against erbb3
WO2013022855A1 (en) 2011-08-05 2013-02-14 Xencor, Inc. Antibodies with modified isoelectric points and immunofiltering
CA2837975C (en) 2011-08-23 2022-04-05 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
MX2014002289A (es) 2011-08-26 2015-03-20 Merrimack Pharmaceuticals Inc Anticuerpos fc especificos en tandem.
US20150050269A1 (en) 2011-09-30 2015-02-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
SG11201401101XA (en) 2011-09-30 2014-08-28 Chugai Pharmaceutical Co Ltd Antigen-binding molecule for promoting loss of antigens
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
EP2766392B1 (en) 2011-10-10 2019-07-17 Xencor, Inc. A method for purifying antibodies
CA2850572A1 (en) 2011-10-20 2013-04-25 Esbatech, A Novartis Company Llc Stable multiple antigen-binding antibody
US11851476B2 (en) 2011-10-31 2023-12-26 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule having regulated conjugation between heavy-chain and light-chain
CN104080811B (zh) 2011-11-04 2019-09-27 酵活有限公司 在Fc结构域中具有突变的稳定异源二聚的抗体设计
US9975956B2 (en) 2011-12-22 2018-05-22 I2 Pharmaceuticals, Inc. Surrogate binding proteins which bind DR4 and/or DR5
ES2694180T3 (es) 2012-01-20 2018-12-18 Vib Vzw Citocinas de haz alfa-helicoidal mutantes dirigidas
TW202015731A (zh) 2012-02-24 2020-05-01 日商中外製藥股份有限公司 經FcγRIIB促進抗原消失之抗原結合分子
NZ772318A (en) 2012-04-20 2023-06-30 Merus Nv Methods and means for the production of ig-like molecules
DK2857419T3 (da) 2012-05-30 2021-03-29 Chugai Pharmaceutical Co Ltd Antigen-bindende molekyle til eliminering af aggregerede antigener
US9499634B2 (en) 2012-06-25 2016-11-22 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
WO2014012085A2 (en) 2012-07-13 2014-01-16 Zymeworks Inc. Bispecific asymmetric heterodimers comprising anti-cd3 constructs
US20140072581A1 (en) 2012-07-23 2014-03-13 Zymeworks Inc. Immunoglobulin Constructs Comprising Selective Pairing of the Light and Heavy Chains
JOP20200236A1 (ar) 2012-09-21 2017-06-16 Regeneron Pharma الأجسام المضادة لمضاد cd3 وجزيئات ربط الأنتيجين ثنائية التحديد التي تربط cd3 وcd20 واستخداماتها
KR20150064068A (ko) 2012-10-08 2015-06-10 로슈 글리카트 아게 2개의 Fab 단편을 포함하는 FC-부재 항체 및 이용 방법
US9562110B2 (en) 2012-11-21 2017-02-07 Wuhan Yzy Biopharma Co., Ltd. Bispecific antibody
US20140377269A1 (en) 2012-12-19 2014-12-25 Adimab, Llc Multivalent antibody analogs, and methods of their preparation and use
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
WO2014113510A1 (en) 2013-01-15 2014-07-24 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
KR20150131208A (ko) 2013-03-13 2015-11-24 이미지냅 인코포레이티드 Cd8에의 항원 결합 구조체들
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
DK2970486T3 (en) 2013-03-15 2018-08-06 Xencor Inc MODULATION OF T-CELLS WITH BISPECIFIC ANTIBODIES AND FC-FUSIONS
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
EP2986312B1 (en) 2013-04-19 2021-12-15 Cytune Pharma Cytokine derived treatment with reduced vascular leak syndrome
WO2014209804A1 (en) 2013-06-24 2014-12-31 Biomed Valley Discoveries, Inc. Bispecific antibodies
US10202433B2 (en) * 2013-06-27 2019-02-12 Inserm (Institut National De La Sante Et De La Recherche Medicale) IL-15 mutant polypeptides as IL-15 antagonists and encoding nucleic acids
CN105593059B (zh) 2013-07-19 2018-07-31 萤火虫医疗公司 用于移动性辅助和输液管理的设备
KR102457731B1 (ko) 2013-08-08 2022-10-21 싸이튠 파마 병용 약학 조성물
EP4269441A3 (en) * 2013-08-08 2024-01-24 Cytune Pharma Il-15 and il-15ralpha sushi domain based on modulokines
EP2839842A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific monovalent diabodies that are capable of binding CD123 and CD3 and uses thereof
SG11201603244VA (en) 2013-11-04 2016-05-30 Glenmark Pharmaceuticals Sa Production of t cell retargeting hetero-dimeric immunoglobulins
CN105899535A (zh) 2013-12-17 2016-08-24 豪夫迈·罗氏有限公司 用pd-1轴结合拮抗剂和抗cd20抗体治疗癌症的方法
ES2811923T3 (es) 2013-12-17 2021-03-15 Genentech Inc Anticuerpos anti-CD3 y métodos de uso
CN106459182B (zh) 2013-12-30 2021-09-03 岸迈生物科技有限公司 串联fab免疫球蛋白及其用途
EP2915569A1 (en) 2014-03-03 2015-09-09 Cytune Pharma IL-15/IL-15Ralpha based conjugates purification method
TWI701042B (zh) 2014-03-19 2020-08-11 美商再生元醫藥公司 用於腫瘤治療之方法及抗體組成物
JP7348708B2 (ja) 2014-04-30 2023-09-21 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ 組み合わせワクチン装置および癌細胞を殺滅する方法
RU2718692C2 (ru) 2014-05-29 2020-04-13 Мэкроудженикс, Инк. Триспецифичные связывающие молекулы, которые специфически связывают антигены множества злокачественных опухолей, и способы их применения
US20150351275A1 (en) 2014-05-30 2015-12-03 Johanson Manufacturing Corporation Thin-Film Radio Frequency Power Terminator
WO2015195163A1 (en) 2014-06-20 2015-12-23 R-Pharm Overseas, Inc. Pd-l1 antagonist fully human antibody
EP3673915A1 (en) 2014-06-30 2020-07-01 Altor BioScience Corporation Il-15-based molecules and methods of use thereof
US20160060360A1 (en) 2014-07-24 2016-03-03 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
JO3663B1 (ar) 2014-08-19 2020-08-27 Merck Sharp & Dohme الأجسام المضادة لمضاد lag3 وأجزاء ربط الأنتيجين
PL3183268T3 (pl) 2014-08-19 2020-09-07 Novartis Ag Chimeryczny receptor antygenowy (CAR) anty-CD123 do zastosowania w leczeniu nowotworu złośliwego
US9763705B2 (en) 2014-10-03 2017-09-19 Globus Medical, Inc. Orthopedic stabilization devices and methods for installation thereof
MX2017004838A (es) 2014-10-14 2017-10-16 Armo Biosciences Inc Composiciones de interleucina-15 y usos de estas.
PL3221355T3 (pl) 2014-11-20 2021-03-08 F. Hoffmann-La Roche Ag Terapia skojarzona składająca się z dwuswoistych aktywujących limfocyty T cząsteczek wiążących antygen CD3 i receptor folianowy 1 (FolR1) oraz antagonistów wiązania osi PD-1
KR20170084326A (ko) 2014-11-26 2017-07-19 젠코어 인코포레이티드 Cd3 및 종양 항원과 결합하는 이종이량체 항체
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
WO2016086186A2 (en) 2014-11-26 2016-06-02 Xencor, Inc. Heterodimeric antibodies including binding to cd8
EP3223907A2 (en) 2014-11-26 2017-10-04 Xencor, Inc. Heterodimeric antibodies that bind cd3 and cd38
SI3235830T1 (sl) 2014-12-19 2020-12-31 Jiangsu Hengrui Medicine Co., Ltd. Proteinski kompleks interlevkina 15 in njegove uporabe
ES2881484T3 (es) 2014-12-22 2021-11-29 Pd 1 Acquisition Group Llc Anticuerpos anti-PD-1
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
EP3064507A1 (en) 2015-03-06 2016-09-07 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Fusion proteins comprising a binding protein and an interleukin-15 polypeptide having a reduced affinity for IL15ra and therapeutic uses thereof
EP4059514A1 (en) 2015-05-08 2022-09-21 Xencor, Inc. Heterodimeric antibodies that bind cd3 and tumor antigens
GEP20227419B (en) 2015-07-30 2022-10-10 Macrogenics Inc Pd-1-binding molecules and methods of use thereof
WO2017053649A1 (en) 2015-09-25 2017-03-30 Altor Bioscience Corporation Interleukin-15 superagonist significantly enhances graft-versus-tumor activity
CN108473578A (zh) 2015-12-22 2018-08-31 瑞泽恩制药公司 治疗癌症的抗pd-1抗体和双特异性抗cd20/抗cd3抗体组合
EP3464370A1 (en) 2016-06-01 2019-04-10 Xencor, Inc. Bispecific antibodies that bind cd20 and cd3 for use in the treatment of lymphoma
US20170349660A1 (en) 2016-06-01 2017-12-07 Xencor. Inc. Bispecific antibodies that bind cd123 and cd3
EP3252078A1 (en) 2016-06-02 2017-12-06 F. Hoffmann-La Roche AG Type ii anti-cd20 antibody and anti-cd20/cd3 bispecific antibody for treatment of cancer
JP2019517539A (ja) 2016-06-07 2019-06-24 マクロジェニクス,インコーポレーテッド 併用療法
EP4257613A3 (en) 2016-06-14 2023-12-13 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
JP6944473B2 (ja) 2016-07-05 2021-10-06 ノバルティス アーゲー 初期サクビトリル中間体のための新規な方法
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
PE20191034A1 (es) 2016-10-14 2019-08-05 Xencor Inc Proteinas de fusion heterodimericas biespecificas que contienen proteinas de fusion fc il-15/il-15r y fragmentos de anticuerpo pd-1
CA3200275A1 (en) 2016-10-21 2018-04-26 Nantcell, Inc. Multimeric il-15-based molecules
US11359018B2 (en) 2016-11-18 2022-06-14 Symphogen A/S Anti-PD-1 antibodies and compositions
EP3645122A1 (en) 2017-06-30 2020-05-06 Xencor, Inc. Targeted heterodimeric fc fusion proteins containing il-15/il-15ra and antigen binding domains
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
EP3749363A1 (en) 2018-02-08 2020-12-16 Amgen Inc. Low ph pharmaceutical antibody formulation
AU2019228381B2 (en) 2018-02-28 2021-12-16 Pfizer Inc. IL-15 variants and uses thereof
AU2019255744A1 (en) 2018-04-18 2020-11-26 Xencor, Inc. IL-15/IL-15Ra heterodimeric Fc fusion proteins and uses thereof
AU2019256539A1 (en) 2018-04-18 2020-11-26 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
CN113423734A (zh) 2018-10-12 2021-09-21 Xencor股份有限公司 靶向PD-1的IL-15/IL-15RαFC融合蛋白及其在联合疗法中的应用
EP3897853A1 (en) 2018-12-20 2021-10-27 Xencor, Inc. Targeted heterodimeric fc fusion proteins containing il-15/il-15ra and nkg2d antigen binding domains
WO2020185739A1 (en) 2019-03-11 2020-09-17 Jounce Therapeutics, Inc. Anti-icos antibodies for the treatment of cancer

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8216805B2 (en) 1995-03-01 2012-07-10 Genentech, Inc. Knobs and holes heteromeric polypeptides
WO1998048032A2 (en) 1997-04-21 1998-10-29 Donlar Corporation POLY-(α-L-ASPARTIC ACID), POLY-(α-L-GLUTAMIC ACID) AND COPOLYMERS OF L-ASP AND L-GLU, METHOD FOR THEIR PRODUCTION AND THEIR USE
US20040214988A1 (en) 2000-03-23 2004-10-28 California Institute Of Technology Method for stabilization of proteins using non-natural amino acids
US6586207B2 (en) 2000-05-26 2003-07-01 California Institute Of Technology Overexpression of aminoacyl-tRNA synthetases for efficient production of engineered proteins containing amino acid analogues
WO2003073238A2 (en) 2002-02-27 2003-09-04 California Institute Of Technology Computational method for designing enzymes for incorporation of amino acid analogs into proteins
WO2005035727A2 (en) 2003-10-09 2005-04-21 Ambrx, Inc. Polymer derivatives
WO2005074524A2 (en) 2004-02-02 2005-08-18 Ambrx, Inc. Modified human interferon polypeptides and their uses
US20120149876A1 (en) 2010-11-05 2012-06-14 Zymeworks Inc. Stable Heterodimeric Antibody Design with Mutations in the Fc Domain
US20140288275A1 (en) 2013-01-14 2014-09-25 Xencor, Inc. Novel heterodimeric proteins
US20140370013A1 (en) 2013-01-14 2014-12-18 Xencor, Inc. Novel heterodimeric proteins
WO2014145806A2 (en) 2013-03-15 2014-09-18 Xencor, Inc. Heterodimeric proteins
AU2014377106A1 (en) * 2014-01-08 2016-08-11 Jiangsu Hengrui Medicine Co., Ltd. IL-15 heterodimeric protein and uses thereof
US20150307629A1 (en) 2014-03-28 2015-10-29 Matthew Bernett Bispecific antibodies that bind to CD38 and CD3
US20160244525A1 (en) 2014-11-05 2016-08-25 Genentech, Inc. Anti-fgfr2/3 antibodies and methods using same
WO2016141387A1 (en) * 2015-03-05 2016-09-09 Xencor, Inc. Modulation of t cells with bispecific antibodies and fc fusions

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
ANDERSON ET AL., PROC NATL ACAD SCI USA, vol. 101, no. 2, 2004, pages 7566 - 71
ATWELL ET AL., J. MOL. BIOL., vol. 270, 1997, pages 26
CHIN ET AL., SCIENCE, vol. 301, no. 5635, 2003, pages 964 - 7
CROPP; SHULTZ, TRENDS GENET., vol. 20, no. 12, 2004, pages 625 - 30
DALL' ACQUA ET AL., J. IMMUNOL., vol. 169, 2002, pages 5171 - 5180
DAVIS ET AL., IMMUNOLOGICAL REVIEWS, vol. 190, 2002, pages 123 - 136
E.A. KABAT: "SEQUENCES OF IMMUNOLOGICAL INTEREST, 5th edition,", NIH
EDELMAN ET AL., PROC NATL ACAD SCI USA, vol. 63, 1969, pages 78 - 85
GHETIE; WARD, IMMUNOL TODAY, vol. 18, no. 12, 1997, pages 592 - 598
GUNASEKARAN ET AL., J. BIOL. CHEM., vol. 285, no. 25, 2010, pages 19637
J. W. CHIN ET AL., JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 124, 2002, pages 9026 - 9027
J. W. CHIN ET AL., PICAS UNITED STATES OF AMERICA, vol. 99, 2002, pages 11020 - 11024
J. W. CHIN; P. G. SCHULTZ, CHEMBIOCHEM, vol. 11, 2002, pages 1135 - 1137
JEFFERIS ET AL., IMMUNOL LETT, vol. 82, 2002, pages 57 - 65
KABAT ET AL.: "Sequences of Proteins of Immunological Interest, 5th Ed.,", 1991, NATIONAL INSTITUTES OF HEALTH
L. WANG; P. G. SCHULTZ, CHEM., vol. 1-10, 2002
MERCHANT ET AL., NATURE BIOTECH., vol. 16, 1998, pages 677
OSOL, A.: "Remington's Pharmaceutical Sciences 16th edition,", 1980
RIDGWAY ET AL., PROTEIN ENGINEERING, vol. 9, no. 7, 1996, pages 617
SIMON ET AL., PNAS USA, vol. 89, no. 20, 1992, pages 9367

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10696722B2 (en) 2016-08-10 2020-06-30 Ajou University Industry-Academic Cooperation Foundation Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
US11078249B2 (en) 2016-08-10 2021-08-03 Ajou University Industry-Academic Cooperation Foundation Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
US11692019B2 (en) 2016-08-10 2023-07-04 Ajou University Industry-Academic Cooperation Foundation Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
US11584794B2 (en) 2016-10-14 2023-02-21 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Ralpha Fc-fusion proteins and immune checkpoint antibody fragments
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-fusion proteins
US10550185B2 (en) 2016-10-14 2020-02-04 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Rα Fc-fusion proteins and PD-1 antibody fragments
US11267883B2 (en) 2016-12-21 2022-03-08 Cephalon, Inc. Antibodies that specifically bind to human IL-15 and uses thereof
WO2019006472A1 (en) * 2017-06-30 2019-01-03 Xencor, Inc. TARGETED HETETRODIMERIC FUSION PROTEINS CONTAINING IL-15 / IL-15RA AND ANTIGEN-BINDING DOMAINS
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
US11059876B2 (en) 2018-02-28 2021-07-13 Pfizer Inc. IL-15 variants and uses thereof
WO2019204592A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof
US11505595B2 (en) 2018-04-18 2022-11-22 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
WO2019204655A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Tim-3 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and tim-3 antigen binding domains
US11524991B2 (en) 2018-04-18 2022-12-13 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
WO2019204665A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Pd-1 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and pd-1 antigen binding domains and uses thereof
WO2019204646A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Lag-3 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and lag-3 antigen binding domains
WO2019229658A1 (en) 2018-05-30 2019-12-05 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
US11377477B2 (en) 2018-10-12 2022-07-05 Xencor, Inc. PD-1 targeted IL-15/IL-15RALPHA fc fusion proteins and uses in combination therapies thereof
CN113286808A (zh) * 2018-10-23 2021-08-20 蜻蜓疗法股份有限公司 异二聚体Fc融合蛋白
US11787864B2 (en) 2018-10-23 2023-10-17 Dragonfly Therapeutics, Inc. Heterodimeric Fc-fused proteins
WO2020086758A1 (en) * 2018-10-23 2020-04-30 Dragonfly Therapeutics, Inc. Heterodimeric fc-fused proteins
WO2020132646A1 (en) * 2018-12-20 2020-06-25 Xencor, Inc. Targeted heterodimeric fc fusion proteins containing il-15/il-15ra and nkg2d antigen binding domains
US11618776B2 (en) 2018-12-20 2023-04-04 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains
US12006345B2 (en) 2019-02-21 2024-06-11 Xencor, Inc. Untargeted and targeted IL-10 Fc-fusion proteins
WO2020172631A3 (en) * 2019-02-21 2020-10-08 Xencor, Inc. Untargeted and targeted il-10 fc-fusion proteins
WO2020231855A1 (en) 2019-05-10 2020-11-19 Nant Holdings Ip, Llc Nogapendekin alfa-inbakicept for immune stimulant therapies and treatment of viral infections
WO2020234387A1 (en) 2019-05-20 2020-11-26 Cytune Pharma IL-2/IL-15Rßy AGONIST DOSING REGIMENS FOR TREATING CANCER OR INFECTIOUS DISEASES
KR20220012296A (ko) 2019-05-20 2022-02-03 싸이튠 파마 암 또는 감염성 질환 치료를 위한 IL-2/IL-15Rβγ 작용제 투여 요법
WO2021006875A1 (en) 2019-07-08 2021-01-14 Nantkwest, Inc. Mononuclear cell derived nk cells
US11453862B2 (en) 2019-07-08 2022-09-27 Immunitybio, Inc. Mononuclear cell derived NK cells
WO2021022304A2 (en) 2019-07-30 2021-02-04 Qlsf Biotherapeutics, Inc. MULTISPECIFIC BINDING COMPOUNDS THAT BIND TO LRRC15 AND CD3ε
WO2021053559A1 (en) 2019-09-18 2021-03-25 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2021054867A1 (ru) 2019-09-19 2021-03-25 Закрытое Акционерное Общество "Биокад" Иммуноцитокин, включающий гетеро димерный белковый комплекс на основе il-15 и il-15rα
WO2021072298A1 (en) 2019-10-11 2021-04-15 Genentech, Inc. Pd-1 targeted il-15/il-15ralpha fc fusion proteins with improved properties
US11932675B2 (en) 2019-10-11 2024-03-19 Genentech, Inc. PD-1 targeted IL-15/IL-15Rα Fc fusion proteins with improved properties
WO2021155042A1 (en) * 2020-01-28 2021-08-05 Genentech, Inc. Il15/il15r alpha heterodimeric fc-fusion proteins for the treatment of cancer
EP4119571A4 (en) * 2020-03-12 2024-04-03 SL Metagen NEW BISPECIFIC PROTEIN AND USE THEREOF
US11365233B2 (en) 2020-04-10 2022-06-21 Cytomx Therapeutics, Inc. Activatable cytokine constructs and related compositions and methods
EP4155316A4 (en) * 2020-05-18 2024-05-08 Shandong Simcere Biopharmaceutical Co., Ltd. HUMAN IL-15 MUTANT AND ITS USE
US11542308B2 (en) 2020-08-07 2023-01-03 Genentech, Inc. FLT3 ligand fusion proteins and methods of use
WO2022031876A1 (en) * 2020-08-07 2022-02-10 Genentech, Inc. Flt3 ligand fusion proteins and methods of use
KR20230096047A (ko) 2020-10-26 2023-06-29 싸이튠 파마 비흑색종 피부암 치료용 IL-2/IL-15Rβγ 작용제
WO2022090203A1 (en) 2020-10-26 2022-05-05 Cytune Pharma IL-2/IL-15Rβү AGONIST FOR TREATING SQUAMOUS CELL CARCINOMA
KR20230096049A (ko) 2020-10-26 2023-06-29 싸이튠 파마 편평 세포 암종 치료용 IL-2/IL-15Rβγ 작용제
WO2022090202A1 (en) 2020-10-26 2022-05-05 Cytune Pharma IL-2/IL-15RBβү AGONIST FOR TREATING NON-MELANOMA SKIN CANCER
US11667687B2 (en) 2021-03-16 2023-06-06 Cytomx Therapeutics, Inc. Masked activatable interferon constructs
WO2022232321A1 (en) * 2021-04-28 2022-11-03 Minotaur Therapeutics, Inc. Humanized chimeric bovine antibodies and methods of use
WO2022268983A3 (en) * 2021-06-23 2023-04-06 Cytune Pharma Interleukin-15 based immunocytokines
WO2022268991A1 (en) 2021-06-23 2022-12-29 Cytune Pharma Interleukin 15 variants
WO2023017191A1 (en) 2021-08-13 2023-02-16 Cytune Pharma Il-2/il-15rbetagamma agonist combination with antibody-drug conjugates for treating cancer
WO2023086772A1 (en) 2021-11-12 2023-05-19 Xencor, Inc. Bispecific antibodies that bind to b7h3 and nkg2d
WO2024011179A1 (en) 2022-07-07 2024-01-11 Genentech, Inc. Combinations of il15/il15r alpha heterodimeric fc-fusion proteins and fcrh5xcd3 bispecific antibodies for the treatment of blood cancers
WO2024092038A2 (en) 2022-10-25 2024-05-02 Ablexis, Llc Anti-cd3 antibodies
WO2024102636A1 (en) 2022-11-07 2024-05-16 Xencor, Inc. Bispecific antibodies that bind to b7h3 and mica/b
WO2024107731A2 (en) 2022-11-14 2024-05-23 Ablexis, Llc Anti-pd-l1 antibodies

Also Published As

Publication number Publication date
KR20190060821A (ko) 2019-06-03
BR112019007281A2 (pt) 2019-07-09
CO2019003945A2 (es) 2019-04-30
EP3526241A1 (en) 2019-08-21
JP7142630B2 (ja) 2022-09-27
AU2022203782B2 (en) 2024-02-22
IL312989A (en) 2024-07-01
SG11201903304YA (en) 2019-05-30
US20200040083A1 (en) 2020-02-06
US10501543B2 (en) 2019-12-10
RU2019114175A (ru) 2020-11-16
RU2019114268A (ru) 2020-11-16
MX2023008559A (es) 2023-08-08
US20220195048A1 (en) 2022-06-23
MA46534A (fr) 2019-08-21
CN110214148A (zh) 2019-09-06
AU2017342560A1 (en) 2019-05-02
CL2019001000A1 (es) 2020-03-27
CL2021003547A1 (es) 2022-09-20
EP3526240A1 (en) 2019-08-21
AU2024203151A1 (en) 2024-06-13
CN110214147A (zh) 2019-09-06
RU2019114268A3 (es) 2021-02-19
BR112019007288A2 (pt) 2019-07-09
CO2019003943A2 (es) 2019-04-30
CA3039930A1 (en) 2018-04-19
US20180118805A1 (en) 2018-05-03
ZA201902380B (en) 2020-08-26
CA3040504A1 (en) 2018-04-19
PH12019500781A1 (en) 2019-06-03
JP2022180450A (ja) 2022-12-06
IL265998B1 (en) 2024-07-01
WO2018071918A1 (en) 2018-04-19
US10550185B2 (en) 2020-02-04
CL2019001001A1 (es) 2020-03-27
AU2022203820A1 (en) 2022-06-23
US20180118828A1 (en) 2018-05-03
KR20240038176A (ko) 2024-03-22
JP2023106405A (ja) 2023-08-01
MX2019004327A (es) 2019-10-14
CR20190227A (es) 2019-08-29
AU2017342559B2 (en) 2022-03-24
JP2019533443A (ja) 2019-11-21
AU2022203782A1 (en) 2022-06-23
KR102649972B1 (ko) 2024-03-22
JP2019533444A (ja) 2019-11-21
ZA201902383B (en) 2020-08-26
US20200123259A1 (en) 2020-04-23
PH12019500782A1 (en) 2019-06-03
CR20230179A (es) 2023-06-12
JP7273453B2 (ja) 2023-05-15
US20230220081A1 (en) 2023-07-13
SG11201903302UA (en) 2019-05-30
PE20191034A1 (es) 2019-08-05
MX2019004328A (es) 2019-09-18
AU2017342560B2 (en) 2022-03-17
IL265997A (en) 2019-06-30
SA519401562B1 (ar) 2023-02-14
KR20190065346A (ko) 2019-06-11
RU2019114175A3 (es) 2021-02-19
CL2022000097A1 (es) 2022-10-14
MA46533A (fr) 2019-08-21
IL265998A (en) 2019-06-30
US11584794B2 (en) 2023-02-21
PE20191033A1 (es) 2019-08-05
AU2017342559A1 (en) 2019-05-02
KR102562519B1 (ko) 2023-08-02

Similar Documents

Publication Publication Date Title
AU2022203782B2 (en) IL15/IL15Ralpha heterodimeric Fc-fusion proteins
US12006345B2 (en) Untargeted and targeted IL-10 Fc-fusion proteins
AU2019256529A1 (en) TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and TIM-3 antigen binding domains
AU2019256520A1 (en) LAG-3 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and LAG-3 antigen binding domains
US11512122B2 (en) IL-7-FC-fusion proteins
US20230146665A1 (en) Il-18-fc fusion proteins
WO2022140701A1 (en) Icos targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and icos antigen binding domains
RU2779938C2 (ru) ГЕТЕРОДИМЕРНЫЕ Fc-СЛИТЫЕ БЕЛКИ IL15/IL15Rα

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17804322

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019519632

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3040504

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112019007281

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20197012663

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017342560

Country of ref document: AU

Date of ref document: 20171016

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017804322

Country of ref document: EP

Effective date: 20190514

ENP Entry into the national phase

Ref document number: 112019007281

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20190410

WWE Wipo information: entry into national phase

Ref document number: 519401562

Country of ref document: SA